1
|
Blanton LS. Murine Typhus: A Review of a Reemerging Flea-Borne Rickettsiosis with Potential for Neurologic Manifestations and Sequalae. Infect Dis Rep 2023; 15:700-716. [PMID: 37987401 PMCID: PMC10660532 DOI: 10.3390/idr15060063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023] Open
Abstract
Murine typhus is an acute febrile illness caused by Rickettsia typhi, an obligately intracellular Gram-negative coccobacillus. Rats (Rattus species) and their fleas (Xenopsylla cheopis) serve as the reservoir and vector of R. typhi, respectively. Humans become infected when R. typhi-infected flea feces are rubbed into flea bite wounds or onto mucous membranes. The disease is endemic throughout much of the world, especially in tropical and subtropical seaboard regions where rats are common. Murine typhus is reemerging as an important cause of febrile illness in Texas and Southern California, where an alternate transmission cycle likely involves opossums (Didelphis virginiana) and cat fleas (Ctenocephalides felis). Although primarily an undifferentiated febrile illness, a range of neurologic manifestations may occur, especially when treatment is delayed. Serology is the mainstay of diagnostic testing, but confirmation usually requires demonstrating seroconversion or a fourfold increase in antibody titer from acute- and convalescent-phase sera (antibodies are seldom detectable in the first week of illness). Thus, early empiric treatment with doxycycline, the drug of choice, is imperative. The purpose of this review is to highlight murine typhus as an important emerging and reemerging infectious disease, review its neurologic manifestations, and discuss areas in need of further study.
Collapse
Affiliation(s)
- Lucas S Blanton
- Department Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
2
|
Londoño AF, Scorpio DG, Dumler JS. Innate immunity in rickettsial infections. Front Cell Infect Microbiol 2023; 13:1187267. [PMID: 37228668 PMCID: PMC10203653 DOI: 10.3389/fcimb.2023.1187267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
Rickettsial agents are a diverse group of alpha-proteobacteria within the order Rickettsiales, which possesses two families with human pathogens, Rickettsiaceae and Anaplasmataceae. These obligate intracellular bacteria are most frequently transmitted by arthropod vectors, a first step in the pathogens' avoidance of host cell defenses. Considerable study of the immune responses to infection and those that result in protective immunity have been conducted. Less study has focused on the initial events and mechanism by which these bacteria avoid the innate immune responses of the hosts to survive within and propagate from host cells. By evaluating the major mechanisms of evading innate immunity, a range of similarities among these bacteria become apparent, including mechanisms to escape initial destruction in phagolysosomes of professional phagocytes, those that dampen the responses of innate immune cells or subvert signaling and recognition pathways related to apoptosis, autophagy, proinflammatory responses, and mechanisms by which these microbes attach to and enter cells or those molecules that trigger the host responses. To illustrate these principles, this review will focus on two common rickettsial agents that occur globally, Rickettsia species and Anaplasma phagocytophilum.
Collapse
Affiliation(s)
- Andrés F. Londoño
- The Henry M. Jackson Foundation for Advancement in Military Medicine, Bethesda, MD, United States
- Department of Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Diana G. Scorpio
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - J. Stephen Dumler
- Department of Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
3
|
Cente M, Danchenko M, Skultety L, Filipcik P, Sekeyova Z. Rickettsia Deregulates Genes Coding for the Neurotoxic Cell Response Pathways in Cerebrocortical Neurons In Vitro. Cells 2023; 12:cells12091235. [PMID: 37174635 PMCID: PMC10177168 DOI: 10.3390/cells12091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Rickettsial infections of the central nervous system (CNS) are manifested by severe neurological symptoms and represent a serious life-threatening condition. Despite the considerable health danger, only a few studies have been conducted focusing on the pathogenesis induced by Rickettsia sp. in CNS. To investigate the signaling pathways associated with the neurotoxic effects of rickettsiae, we employed an experimental model of cerebrocortical neurons combined with molecular profiling and comprehensive bioinformatic analysis. The cytopathic effect induced by Rickettsia akari and Rickettsia slovaca was demonstrated by decreased neuronal viability, structural changes in cell morphology, and extensive fragmentation of neurites in vitro. Targeted profiling revealed the deregulation of genes involved in the neuroinflammatory and neurotoxic cell response pathways. Although quantitative analysis showed differences in gene expression response, functional annotation revealed that the biological processes are largely shared between both Rickettsia species. The identified enriched pathways are associated with cytokine signaling, chemotaxis of immune cells, responses to infectious agents, interactions between neurons, endothelial and glial cells, and regulation of neuronal apoptotic processes. The findings of our study provide new insight into the etiopathogenesis of CNS infection and further expand the understanding of molecular signaling associated with neuroinvasive Rickettsia species.
Collapse
Affiliation(s)
- Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Monika Danchenko
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Ludovit Skultety
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Zuzana Sekeyova
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
4
|
Rickettsia Vaccine Candidate pVAX1-OmpB24 Stimulates TCD4+INF-γ+ and TCD8+INF-γ+ Lymphocytes in Autologous Co-Culture of Human Cells. Vaccines (Basel) 2023; 11:vaccines11010173. [PMID: 36680017 PMCID: PMC9865178 DOI: 10.3390/vaccines11010173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND In recent years, promising vaccination strategies against rickettsiosis have been described in experimental animal models and human cells. OmpB is considered an immunodominant antigen that is recognized by T and B cells. The aim of this study was to identify TCD4+INF-γ+ and TCD8+INF-γ+ lymphocytes in an autologous system with macrophages transfected with the vaccine candidate pVAX1-OmpB24. Lymphocytes and monocytes from 14 patients with Rickettsia were isolated from whole blood. Monocytes were differentiated into macrophages and transfected with the plasmid pVAX1-OmpB24 pVax1. Isolated lymphocytes were cultured with transfected macrophages. IFN-γ-producing TCD4+ and TCD8+ lymphocyte subpopulations were identified by flow cytometry, as was the percentage of macrophages expressing CD40+, CD80+, HLA-I and HLA-II. Also, we analyzed the exhausted condition of the T lymphocyte subpopulation by PD1 expression. Macrophages transfected with pVAX1-OmpB24 stimulated TCD4+INF-γ+ cells in healthy subjects and patients infected with R. typhi. Macrophages stimulated TCD8+INF-γ+ cells in healthy subjects and patients infected with R. rickettsii and R. felis. Cells from healthy donors stimulated with OmpB-24 showed a higher percentage of TCD4+PD1+. Cells from patients infected with R. rickettsii had a higher percentage of TCD8+PD-1+, and for those infected with R. typhi the larger number of cells corresponded to TCD4+PD1+. Human macrophages transfected with pVAX1-OmpB24 activated TCD4+IFN-γ+ and CD8+IFN-γ+ in patients infected with different Rickettsia species. However, PD1 expression played an important role in the inhibition of T lymphocytes with R. felis.
Collapse
|
5
|
IL-1 Superfamily Member ( IL-1A, IL-1B and IL-18) Genetic Variants Influence Susceptibility and Clinical Course of Mediterranean Spotter Fever. Biomolecules 2022; 12:biom12121892. [PMID: 36551320 PMCID: PMC9816934 DOI: 10.3390/biom12121892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/01/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Mediterranean Spotted Fever (MSF) is one of the most common spotted fever Rickettsioses. Most cases of MSF follow a benign course, with a minority of cases being fatal. The severity of the infection depends on bacterial virulence, dose and host factors such as effective immune response and genetic background. Herein, we reported data on typing by competitive allele-specific PCR of functionally relevant polymorphisms of genes coding for MyD88 adapter-like (Mal/TIRAP) protein (rs8177374), interleukin(IL)-1 cluster (IL-1A rs1800587, IL-1B rs16944 and rs1143634) and IL-18 (rs187238), which might be crucial for an efficient immune response. The results enlighten the role that IL-1 gene cluster variants might play in susceptibility against Rickettsia conorii infection. In particular, the IL-1A rs1800587TT genotype was significantly increased in patients alone and combined in a haplotype composed by minor alleles rs1800587T, rs16944A and rs1143634A. This result was confirmed using the decision tree heuristic approach. Using this methodology, IL-1A rs1800587TT genotype was the better discrimination key among MSF patients and controls. In addition, the IL-1 gene cluster SNP genotypes containing minor alleles and IL-18 rs187238G positive genotypes were found as associated with risk of severe complications such as sepsis, septic shock, acute respiratory distress syndrome and coma. In conclusion, these data suggest that the evaluation of IL-1A, IL-1B and IL-18 gene SNPs can add useful information on the clinical course of patients affected by Mediterranean Spotted Fever, even if further confirmatory studies will be necessary.
Collapse
|
6
|
Involvement of Pore Formation and Osmotic Lysis in the Rapid Killing of Gamma Interferon-Pretreated C166 Endothelial Cells by Rickettsia prowazekii. Trop Med Infect Dis 2022; 7:tropicalmed7080163. [PMID: 36006255 PMCID: PMC9415803 DOI: 10.3390/tropicalmed7080163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Rickettsia prowazekii, the bacterial cause of epidemic typhus in humans, proliferates mainly within the microvascular endothelial cells. Previous studies have shown that murine macrophage-like RAW264.7 cells are rapidly damaged if they are pretreated with gamma interferon (IFN-γ) and then infected with R. prowazekii. In the present study, the effects of IFN-γ and R. prowazekii on murine C166 endothelial cells were evaluated. In the IFN-γ-pretreated R. prowazekii-infected endothelial cell cultures, evidence of cell damage was observed within several hours after addition of the rickettsiae. Considerable numbers of the cells became permeable to trypan blue dye and ethidium bromide, and substantial amounts of lactate dehydrogenase (LDH) were released from the cells. Such evidence of cellular injury was not observed in the untreated infected cultures or in any of the mock-infected cultures. Polyethylene glycols (PEGs) of different nominal average molecular weights were used to assess the possible involvement of pore formation and osmotic lysis in this cellular injury. PEG 8000 dramatically suppressed LDH release, PEG 4000 partially inhibited it, and PEGs 2000 and 1450 had no effect. Despite its inhibition of LDH release, PEG 8000 did not prevent the staining of the IFN-γ-pretreated infected endothelial cells by ethidium bromide. These findings suggest that the observed cellular injury involves the formation of pores in the endothelial cell membranes, followed by osmotic lysis of the cells.
Collapse
|
7
|
Rauch J, Jochum J, Eisermann P, Gisbrecht J, Völker K, Hunstig F, Mehlhoop U, Muntau B, Tappe D. Inflammatory cytokine profile and T cell responses in African tick bite fever patients. Med Microbiol Immunol 2022; 211:143-152. [PMID: 35543881 PMCID: PMC9092931 DOI: 10.1007/s00430-022-00738-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Abstract
African tick bite fever, an acute febrile illness, is caused by the obligate intracellular bacterium Rickettsia africae. Immune responses to rickettsial infections have so far mainly been investigated in vitro with infected endothelial cells as the main target cells, and in mouse models. Patient studies are rare and little is known about the immunology of human infections. In this study, inflammatory mediators and T cell responses were examined in samples from 13 patients with polymerase chain reaction-confirmed R. africae infections at different time points of illness. The Th1-associated cytokines IFNγ and IL-12 were increased in the acute phase of illness, as were levels of the T cell chemoattractant cytokine CXCL-10. In addition, the anti-inflammatory cytokine IL-10 and also IL-22 were elevated. IL-22 but not IFNγ was increasingly produced by CD4+ and CD8+ T cells during illness. Besides IFNγ, IL-22 appears to play a protective role in rickettsial infections.
Collapse
Affiliation(s)
- Jessica Rauch
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany.
| | - Johannes Jochum
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philip Eisermann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Jana Gisbrecht
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | | | | | - Ute Mehlhoop
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Birgit Muntau
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| |
Collapse
|
8
|
Dahmani M, Cook JH, Zhu JC, Riley SP. Contribution of classical complement activation and IgM to the control of Rickettsia infection. Mol Microbiol 2021; 116:1476-1488. [PMID: 34725868 PMCID: PMC8955150 DOI: 10.1111/mmi.14839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/02/2023]
Abstract
Pathogenic Rickettsia are obligate intracellular bacteria and the etiologic agents of many life‐threatening infectious diseases. Due to the serious nature of these infections, it is imperative to both identify the responsive immune sensory pathways and understand the associated immune mechanisms that restrict Rickettsia proliferation. Previous studies have demonstrated that the mammalian complement system is both activated during Rickettsia infection and contributes to the immune response to infection. To further define this component of the mammalian anti‐Rickettsia immune response, we sought to identify the mechanism(s) of complement activation during Rickettsia infection. We have employed a series of in vitro and in vivo models of infection to investigate the role of the classical complement activation pathway during Rickettsia infection. Depletion or elimination of complement activity demonstrates that both C1q and pre‐existing IgM contribute to complement activation; thus implicating the classical complement system in Rickettsia‐mediated complement activation. Elimination of the classical complement pathway from mice increases susceptibility to R. australis infection with both increased bacterial loads in multiple tissues and decreased immune activation markers. This study highlights the role of the classical complement pathway in immunity against Rickettsia and implicates resident Rickettsia‐responsive IgM in the response to infection.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jack H Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jinyi C Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Sean P Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
9
|
Vaccine Design and Vaccination Strategies against Rickettsiae. Vaccines (Basel) 2021; 9:vaccines9080896. [PMID: 34452021 PMCID: PMC8402588 DOI: 10.3390/vaccines9080896] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/30/2022] Open
Abstract
Rickettsioses are febrile, potentially lethal infectious diseases that are a serious health threat, especially in poor income countries. The causative agents are small obligate intracellular bacteria, rickettsiae. Rickettsial infections are emerging worldwide with increasing incidence and geographic distribution. Nonetheless, these infections are clearly underdiagnosed because methods of diagnosis are still limited and often not available. Another problem is that the bacteria respond to only a few antibiotics, so delayed or wrong antibiotic treatment often leads to a more severe outcome of the disease. In addition to that, the development of antibiotic resistance is a serious threat because alternative antibiotics are missing. For these reasons, prophylactic vaccines against rickettsiae are urgently needed. In the past years, knowledge about protective immunity against rickettsiae and immunogenic determinants has been increasing and provides a basis for vaccine development against these bacterial pathogens. This review provides an overview of experimental vaccination approaches against rickettsial infections and perspectives on vaccination strategies.
Collapse
|
10
|
Caravedo Martinez MA, Ramírez-Hernández A, Blanton LS. Manifestations and Management of Flea-Borne Rickettsioses. Res Rep Trop Med 2021; 12:1-14. [PMID: 33574726 PMCID: PMC7873028 DOI: 10.2147/rrtm.s274724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Murine typhus and flea-borne spotted fever are undifferentiated febrile illnesses caused by Rickettsia typhi and Rickettsia felis, respectively. These organisms are small obligately intracellular bacteria and are transmitted to humans by fleas. Murine typhus is endemic to coastal areas of the tropics and subtropics (especially port cities), where rats are the primary mammalian host and rat fleas (Xenopsylla cheopis) are the vector. In the United States, a cycle of transmission involving opossums and cat fleas (Ctenocephalides felis) are the presumed reservoir and vector, respectively. The incidence and distribution of murine typhus appear to be increasing in endemic areas of the US. Rickettsia felis has also been reported throughout the world and is found within the ubiquitous cat flea. Flea-borne rickettsioses manifest as an undifferentiated febrile illness. Headache, malaise, and myalgia are frequent symptoms that accompany fever. The incidence of rash is variable, so its absence should not dissuade the clinician to consider a rickettsial illness as part of the differential diagnosis. When present, the rash is usually macular or papular. Although not a feature of murine typhus, eschar has been found in 12% of those with flea-borne spotted fever. Confirmatory laboratory diagnosis is usually obtained by serology; the indirect immunofluorescence assay is the serologic test of choice. Antibodies are seldom present during the first few days of illness. Thus, the diagnosis requires acute- and convalescent-phase specimens to document seroconversion or a four-fold increase in antibody titer. Since laboratory diagnosis is usually retrospective, when a flea-borne rickettsiosis is considered, empiric treatment should be initiated. The treatment of choice for both children and adults is doxycycline, which results in a swift and effective response. The following review is aimed to summarize the key clinical, epidemiological, ecological, diagnostic, and treatment aspects of flea-borne rickettsioses.
Collapse
Affiliation(s)
- Maria A Caravedo Martinez
- Department of Internal Medicine – Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Lucas S Blanton
- Department of Internal Medicine – Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
11
|
Blanton LS, Berman MA, Afrouzian M. Case Report: Renal Failure due to Focal Segmental Glomerulosclerosis in a Patient with Murine Typhus. Am J Trop Med Hyg 2020; 103:1017-1019. [PMID: 32588799 DOI: 10.4269/ajtmh.20-0116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Murine typhus is a flea-borne rickettsiosis caused by Rickettsia typhi. When severe, endothelial dysfunction can lead to acute kidney injury secondary to prerenal azotemia or acute tubular necrosis. Here, we describe an unusual cause of kidney injury during the course of murine typhus-focal segmental glomerulosclerosis.
Collapse
Affiliation(s)
- Lucas S Blanton
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Megan A Berman
- Division of General Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Marjan Afrouzian
- Division of General Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
12
|
Abstract
Over the last decades, rickettsioses are emerging worldwide. These diseases are caused by intracellular bacteria. Although rickettsioses can be treated with antibiotics, a vaccine against rickettsiae is highly desired for several reasons. Rickettsioses are highly prevalent, especially in poor countries, and there are indications of the development of antibiotic resistance. In addition, some rickettsiae can persist and cause recurrent disease. The development of a vaccine requires the understanding of the immune mechanisms that are involved in protection as well as in immunopathology. Knowledge about these immune responses is accumulating, and efforts have been undertaken to identify antigenic components of rickettsiae that may be useful as a vaccine. This review provides an overview on current knowledge of adaptive immunity against rickettsiae, which is essential for defense, rickettsial antigens that have been identified so far, and on vaccination strategies that have been used in animal models of rickettsial infections.
Collapse
|
13
|
Sarathy VV, Walker DH. Ideal Criteria for Accurate Mouse Models of Vector-Borne Diseases with Emphasis on Scrub Typhus and Dengue. Am J Trop Med Hyg 2020; 103:970-975. [PMID: 32602433 PMCID: PMC7470543 DOI: 10.4269/ajtmh.19-0955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/05/2020] [Indexed: 11/07/2022] Open
Abstract
Nine criteria regarding the infectious agent, mode of transmission, portal of entry, route of spread, target organs, target cells, pathologic lesions, incubation period, and modifiable spectrum of disease and outcomes appropriate to the intended experimental purpose are described. To provide context for each criterion, mouse models of two vector-borne zoonotic infectious diseases, scrub typhus and dengue, are summarized. Application of the criteria indicates that intravenous inoculation of Orientia tsutsugamushi into inbred mice is the best current model for life-threatening scrub typhus, and intradermal inoculation accurately models sublethal human scrub typhus, whereas the immunocompromised mouse models of dengue provide disease outcomes most closely associated with human dengue. In addition to addressing basic questions of immune and pathogenic mechanisms, mouse models are useful for preclinical testing of experimental vaccines and therapeutics. The nine criteria serve as guidelines to evaluate and compare models of vector-borne infectious diseases.
Collapse
Affiliation(s)
- Vanessa V. Sarathy
- Department of Pathology, Sealy Institute for Vaccine Sciences, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - David H. Walker
- Department of Pathology, Sealy Institute for Vaccine Sciences, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
14
|
Narra HP, Sahni A, Walker DH, Sahni SK. Recent research milestones in the pathogenesis of human rickettsioses and opportunities ahead. Future Microbiol 2020; 15:753-765. [PMID: 32691620 PMCID: PMC7787141 DOI: 10.2217/fmb-2019-0266] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/14/2020] [Indexed: 12/27/2022] Open
Abstract
Infections caused by pathogenic Rickettsia species continue to scourge human health across the globe. From the point of entry at the site of transmission by arthropod vectors, hematogenous dissemination of rickettsiae occurs to diverse host tissues leading to 'rickettsial vasculitis' as the salient feature of pathogenesis. This perspective article accentuates recent breakthrough developments in the context of host-pathogen-vector interactions during rickettsial infections. The subtopics include potential exploitation of circulating macrophages for spread, identification of new entry mechanisms and regulators of actin-based motility, appreciation of metabolites acquired from and effectors delivered into the host, importance of the toxin-antitoxin module in host-cell interactions, effects of the vector microbiome on rickettsial transmission, and niche-specific riboregulation and adaptation. Further research on these aspects will advance our understanding of the biology of rickettsiae as intracellular pathogens and should enable design and development of new approaches to counter rickettsioses in humans and other hosts.
Collapse
Affiliation(s)
- Hema P Narra
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Abha Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - David H Walker
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sanjeev K Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
15
|
Quantitative Proteomics of the Endothelial Secretome Identifies RC0497 as Diagnostic of Acute Rickettsial Spotted Fever Infections. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:306-322. [PMID: 31955791 DOI: 10.1016/j.ajpath.2019.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 12/29/2022]
Abstract
Mediterranean spotted fever is a reemerging acute tick-borne infection produced by the α-proteobacterium, Rickettsia conorii. Rickettsia conorii infects vascular endothelial cells producing disseminated plasma leakage, manifesting as nonspecific fever, headache, and maculopapular rash. Because there are no available tests of early infection, Mediterranean spotted fever is often undiagnosed and untreated, resulting in significant mortality. To address this critical need, we have applied a quantitative proteomics pipeline for analyzing the secretome of primary human umbilical vein endothelial cells. Of the 104 proteins whose abundance changed significantly in the R. conorii-infected human umbilical vein endothelial cells' secretome, 46 proteins were up-regulated: 45 were host secreted proteins (including cytokines), and 1 was a rickettsial protein, the putative N-acetylmuramoyl-l-alanine amidase RC0497. Proteins with sequence highly homologous to RC0497 were found to be shared by many species of the spotted fever group rickettsiae, but not typhus group rickettsiae. Quantitative targeted proteomics studies of plasma from a mouse model of sublethal and lethal R. conorii identified RC0497 in the blood, and its circulating levels were proportionally associated with infection outcome. Finally, the presence of RC0497 in the serum samples from a cohort of humans presenting with acute rickettsioses was confirmed. The detection of RC0497 has the potential to be a sensitive and specific marker for acute rickettsial spotted rickettsioses.
Collapse
|
16
|
Londoño AF, Mendell NL, Walker DH, Bouyer DH. A biosafety level-2 dose-dependent lethal mouse model of spotted fever rickettsiosis: Rickettsia parkeri Atlantic Rainforest strain. PLoS Negl Trop Dis 2019; 13:e0007054. [PMID: 31216274 PMCID: PMC6602283 DOI: 10.1371/journal.pntd.0007054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 07/01/2019] [Accepted: 05/21/2019] [Indexed: 11/28/2022] Open
Abstract
Background The species of the Rickettsia genus are separated into four groups: the ancestral group, typhus group, transitional group and spotted fever group. Rickettsia parkeri, a spotted fever group Rickettsia, has been reported across the American continents as infecting several tick species and is associated with a relatively mild human disease characterized by eschar formation at the tick feeding site, regional lymphadenopathy, fever, myalgia and rash. Currently, there are several mouse models that provide good approaches to study the acute lethal disease caused by Rickettsia, but these models can only be performed in an animal biosafety level 3 laboratory. We present an alternative mouse model for acute lethal rickettsial disease, using R. parkeri Atlantic Rainforest strain and C3H/HeN mice, with the advantage that this model can be studied in an animal biosafety level 2 laboratory. Principal findings In the C3H/HeN mouse model, we determined that infection with 1x106 and 1x107 viable R. parkeri Atlantic Rainforest strain organisms produced dose-dependent severity, whereas infection with 1x108 viable bacteria resulted in a lethal illness. The animals became moribund on day five or six post-infection. The lethal disease was characterized by ruffled fur, erythema, labored breathing, decreased activity, and hunched posture, which began on day three post-infection (p.i.) and coincided with the peak bacterial loads. Significant splenomegaly (on days three and five p.i.), neutrophilia (on days three and five p.i.), and thrombocytopenia (on days one, three and five p.i.) were observed. Significance Since R. parkeri is used at biosafety level 2, the greatest advantage of this inbred mouse model is the ability to investigate immunity and pathogenesis of rickettsiosis with all the tools available at biosafety level 2. Rickettsia is a bacterial genus that contains distinct species that are transmitted by arthropods. Many of these agents produce infection and disease in humans. The illness can range from very severe, such as Rocky Mountain spotted fever caused by Rickettsia rickettsii to mild human disease characterized by eschar formation at the tick feeding site and less severe symptoms caused by Rickettsia parkeri and often apparently asymptomatic seroconversion as observed with R. amblyommatis. To study these diseases, animal models are invaluable, and mouse models offer the best advantages for studies of immunity and pathogenesis because of the availability of immunologic reagents and gene knockout animals. Several mouse models are available for the study of the acute lethal disease produced by these bacteria, providing the opportunity to test different treatments and vaccine candidates. However, work with these models requires an animal biosafety level 3 laboratory. In this report, we present an alternative mouse model with R. parkeri Atlantic Rainforest strain available for investigation in a biosafety level 2 laboratory to study an acute dose-dependent lethal spotted fever group rickettsial disease with the advantage that experiments can be performed at this biosafety level.
Collapse
Affiliation(s)
- Andrés F. Londoño
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Research Group on Veterinary Sciences “Centauro”, Facultad de Ciencias Agrarias, Universidad de Antioquia, Medellín, Antioquia, Colombia
- * E-mail:
| | - Nicole L. Mendell
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David H. Walker
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H. Bouyer
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
17
|
Rauch J, Eisermann P, Noack B, Mehlhoop U, Muntau B, Schäfer J, Tappe D. Typhus Group Rickettsiosis, Germany, 2010-2017 1. Emerg Infect Dis 2019; 24:1213-1220. [PMID: 29912688 PMCID: PMC6038764 DOI: 10.3201/eid2407.180093] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Typhus group rickettsiosis is caused by the vectorborne bacteria Rickettsia typhi and R. prowazekii. R. typhi, which causes murine typhus, the less severe endemic form of typhus, is transmitted by fleas; R. prowazekii, which causes the severe epidemic form of typhus, is transmitted by body lice. To examine the immunology of human infection with typhus group rickettsiae, we retrospectively reviewed clinical signs and symptoms, laboratory changes, and travel destinations of 28 patients who had typhus group rickettsiosis diagnosed by the German Reference Center for Tropical Pathogens, Hamburg, Germany, during 2010-2017. Immunofluorescence assays of follow-up serum samples indicated simultaneous seroconversion of IgM, IgA, and IgG or concurrence in the first serum sample. Cytokine levels peaked during the second week of infection, coinciding with organ dysfunction and seroconversion. For 3 patients, R. typhi was detected by species-specific nested quantitative PCR. For all 28 patients, R. typhi was the most likely causative pathogen.
Collapse
|
18
|
Sahni A, Fang R, Sahni SK, Walker DH. Pathogenesis of Rickettsial Diseases: Pathogenic and Immune Mechanisms of an Endotheliotropic Infection. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:127-152. [PMID: 30148688 DOI: 10.1146/annurev-pathmechdis-012418-012800] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obligately intracytosolic rickettsiae that cycle between arthropod and vertebrate hosts cause human diseases with a spectrum of severity, primarily by targeting microvascular endothelial cells, resulting in endothelial dysfunction. Endothelial cells and mononuclear phagocytes have important roles in the intracellular killing of rickettsiae upon activation by the effector molecules of innate and adaptive immunity. In overwhelming infection, immunosuppressive effects contribute to the severity of illness. Rickettsia-host cell interactions involve host cell receptors for rickettsial ligands that mediate cell adhesion and, in some instances, trigger induced phagocytosis. Rickettsiae interact with host cell actin to effect both cellular entry and intracellular actin-based mobility. The interaction of rickettsiae with the host cell also involves rickettsial evasion of host defense mechanisms and exploitation of the intracellular environment. Signal transduction events exemplify these effects. An intriguing frontier is the array of rickettsial noncoding RNA molecules and their potential effects on the pathogenesis and transmission of rickettsial diseases.
Collapse
Affiliation(s)
- Abha Sahni
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - Rong Fang
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - Sanjeev K Sahni
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| | - David H Walker
- The University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0609, USA; , , ,
| |
Collapse
|
19
|
Immunity against the Obligate Intracellular Bacterial Pathogen Rickettsia australis Requires a Functional Complement System. Infect Immun 2018; 86:IAI.00139-18. [PMID: 29581196 PMCID: PMC5964522 DOI: 10.1128/iai.00139-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/21/2018] [Indexed: 11/23/2022] Open
Abstract
The complement system has a well-defined role in deterring blood-borne infections. However, complement is not entirely efficacious, as several bacterial pathogens, including some obligate intracellular pathogens, have evolved mechanisms for resistance. It is presumed that obligate intracellular bacteria evade complement attack by residing within a host cell; however, recent studies have challenged this presumption. Here, we demonstrate that the complement system is activated during infection with the obligate intracellular bacterium Rickettsia australis and that genetic ablation of complement increases susceptibility to infection. Interaction of Rickettsia australis with serum-borne complement leads to activation of the complement cascade, producing three effector mechanisms that could negatively influence R. australis. The C9-dependent membrane attack complex can lead to deposition of a bacteriolytic membrane pore on the bacteria, but this system does not contribute to control of rickettsial infection. Similarly, complement receptor (CR1/2)-dependent opsonophagocytosis may lead to engulfment and killing of the bacteria, but this system is also dispensable for immunity. Nevertheless, intact complement is essential for naturally acquired and antibody-mediated immunity to Rickettsia infection. Comparison of infection in mice lacking the central complement protein C3 with infection in their wild-type counterparts demonstrated decreases in gamma interferon (IFN-γ) production, IgG secretion, and spleen hyperplasia in animals lacking complement. The correlation between loss of secondary immune functions and loss of complement indicates that the proinflammatory signaling components of the complement system, and not membrane attack complex or opsonophagocytosis, contribute to the immune response to this pathogen.
Collapse
|
20
|
Serum cytokine responses in Rickettsia felis infected febrile children, Ghana. Med Microbiol Immunol 2018; 207:243-248. [PMID: 29736763 PMCID: PMC6096778 DOI: 10.1007/s00430-018-0544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/24/2018] [Indexed: 11/13/2022]
Abstract
The intracellular pathogen Rickettsia felis causes flea-borne spotted fever and is increasingly recognized as an emerging cause of febrile illness in Africa, where co-infection with Plasmodium falciparum is common. Rickettsiae invade endothelial cells. Little is known, however, about the early immune responses to infection. In this study, we characterize for the first time the cytokine profile in the acute phase of illness caused by R. felis infection, as well as in plasmodial co-infection, using serum from 23 febrile children < 15 years of age and 20 age-matched healthy controls from Ghana. Levels of IL-8 (interleukin-8), IP-10 (interferon-γ-induced protein-10), MCP-1 (monocyte chemotactic protein-1), MIP-1α (macrophage inflammatory protein-1α) and VEGF (vascular endothelial growth factor) were significantly elevated in R. felis mono-infection; however, IL-8 and VEGF elevation was not observed in plasmodial co-infections. These results have important implications in understanding the early immune responses to R. felis and suggest a complex interplay in co-infections.
Collapse
|
21
|
Osterloh A. Immune response against rickettsiae: lessons from murine infection models. Med Microbiol Immunol 2017; 206:403-417. [PMID: 28770333 PMCID: PMC5664416 DOI: 10.1007/s00430-017-0514-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022]
Abstract
Rickettsiae are small intracellular bacteria that can cause life-threatening febrile diseases. Rickettsioses occur worldwide with increasing incidence. Therefore, a vaccine is highly desired. A prerequisite for the development of a vaccine is the knowledge of the immune response against these bacteria, in particular protective immunity. In recent years murine models of rickettsial infections have been established, and the study of immune response against rickettsiae in mice provided many new insights into protective and pathological immune reactions. This review summarizes the current knowledge about immune mechanisms in protection and pathology in rickettsial infections.
Collapse
Affiliation(s)
- Anke Osterloh
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
22
|
Cytotoxic effector functions of T cells are not required for protective immunity against fatal Rickettsia typhi infection in a murine model of infection: Role of TH1 and TH17 cytokines in protection and pathology. PLoS Negl Trop Dis 2017; 11:e0005404. [PMID: 28222146 PMCID: PMC5336310 DOI: 10.1371/journal.pntd.0005404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/03/2017] [Accepted: 02/12/2017] [Indexed: 01/05/2023] Open
Abstract
Endemic typhus caused by Rickettsia (R.) typhi is an emerging febrile disease that can be fatal due to multiple organ pathology. Here we analyzed the requirements for protection against R. typhi by T cells in the CB17 SCID model of infection. BALB/c wild-type mice generate CD4+ TH1 and cytotoxic CD8+ T cells both of which are sporadically reactivated in persistent infection. Either adoptively transferred CD8+ or CD4+ T cells protected R. typhi-infected CB17 SCID mice from death and provided long-term control. CD8+ T cells lacking either IFNγ or Perforin were still protective, demonstrating that the cytotoxic function of CD8+ T cells is not essential for protection. Immune wild-type CD4+ T cells produced high amounts of IFNγ, induced the release of nitric oxide in R. typhi-infected macrophages and inhibited bacterial growth in vitro via IFNγ and TNFα. However, adoptive transfer of CD4+IFNγ-/- T cells still protected 30-90% of R. typhi-infected CB17 SCID mice. These cells acquired a TH17 phenotype, producing high amounts of IL-17A and IL-22 in addition to TNFα, and inhibited bacterial growth in vitro. Surprisingly, the neutralization of either TNFα or IL-17A in CD4+IFNγ-/- T cell recipient mice did not alter bacterial elimination by these cells in vivo, led to faster recovery and enhanced survival compared to isotype-treated animals. Thus, collectively these data show that although CD4+ TH1 cells are clearly efficient in protection against R. typhi, CD4+ TH17 cells are similarly protective if the harmful effects of combined production of TNFα and IL-17A can be inhibited.
Collapse
|
23
|
Moderzynski K, Papp S, Rauch J, Heine L, Kuehl S, Richardt U, Fleischer B, Osterloh A. CD4+ T Cells Are as Protective as CD8+ T Cells against Rickettsia typhi Infection by Activating Macrophage Bactericidal Activity. PLoS Negl Trop Dis 2016; 10:e0005089. [PMID: 27875529 PMCID: PMC5119731 DOI: 10.1371/journal.pntd.0005089] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/02/2016] [Indexed: 01/07/2023] Open
Abstract
Rickettsia typhi is an intracellular bacterium that causes endemic typhus, a febrile disease that can be fatal due to complications including pneumonia, hepatitis and meningoencephalitis, the latter being a regular outcome in T and B cell-deficient C57BL/6 RAG1-/- mice upon Rickettsia typhi infection. Here, we show that CD4+ TH1 cells that are generated in C57BL/6 mice upon R. typhi infection are as protective as cytotoxic CD8+ T cells. CD4+- as well as CD8+-deficient C57BL/6 survived the infection without showing symptoms of disease at any point in time. Moreover, adoptively transferred CD8+ and CD4+ immune T cells entered the CNS of C57BL/6 RAG1-/- mice with advanced infection and both eradicated the bacteria. However, immune CD4+ T cells protected only approximately 60% of the animals from death. They induced the expression of iNOS in infiltrating macrophages as well as in resident microglia in the CNS which can contribute to bacterial killing but also accelerate pathology. In vitro immune CD4+ T cells inhibited bacterial growth in infected macrophages which was in part mediated by the release of IFNγ. Collectively, our data demonstrate that CD4+ T cells are as protective as CD8+ T cells against R. typhi, provided that CD4+ TH1 effector cells are present in time to support bactericidal activity of phagocytes via the release of IFNγ and other factors. With regard to vaccination against TG Rickettsiae, our findings suggest that the induction of CD4+ TH1 effector cells is sufficient for protection. Endemic typhus caused by Rickettsia typhi usually is a relatively mild disease. However, CNS inflammation and neurological symptoms are complications that can occur in severe cases. This outcome of disease is regularly observed in T and B cell-deficient C57BL/6 RAG1-/- mice upon infection with R. typhi. We show here that CD4+ T cells are as protective as cytotoxic CD8+ T cells against R. typhi as long as they are present in time. This is evidenced by the fact that neither CD8+ nor CD4+ T cell-deficient C57BL/6 mice develop disease which is also true for R. typhi-infected C57BL/6 RAG1-/- mice that receive immune CD8+ or CD4+ at an early point in time. Moreover, adoptive transfer of immune CD4+ T cells still protects approximately 60% of C57BL/6 RAG1-/- mice when applied later in advanced infection when the bacteria start to rise. Although CD8+ T cells are faster and more efficient in bacterial elimination, R. typhi is not detectable in CD4+ T cell recipients anymore. We further show that immune CD4+ T cells activate bactericidal functions of microglia and macrophages in the CNS in vivo and inhibit bacterial growth in infected macrophages in vitro which is in part mediated by the release of IFNγ. Collectively, we demonstrate for the first time that CD4+ T cells alone are sufficient to protect against R. typhi infection. With regard to vaccination our findings suggest that the induction of R. typhi-specific CD4+ TH1 effector T cells may be as effective as the much more difficult targeting of cytotoxic CD8+ T cells.
Collapse
Affiliation(s)
- Kristin Moderzynski
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stefanie Papp
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jessica Rauch
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Liza Heine
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Svenja Kuehl
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ulricke Richardt
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Bernhard Fleischer
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Osterloh
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- * E-mail:
| |
Collapse
|
24
|
Hauptmann M, Kolbaum J, Lilla S, Wozniak D, Gharaibeh M, Fleischer B, Keller CA. Protective and Pathogenic Roles of CD8+ T Lymphocytes in Murine Orientia tsutsugamushi Infection. PLoS Negl Trop Dis 2016; 10:e0004991. [PMID: 27606708 PMCID: PMC5015871 DOI: 10.1371/journal.pntd.0004991] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/19/2016] [Indexed: 01/12/2023] Open
Abstract
T cells are known to contribute to immune protection against scrub typhus, a potentially fatal infection caused by the obligate intracellular bacterium Orientia (O.) tsutsugamushi. However, the contribution of CD8+ T cells to protection and pathogenesis during O. tsutsugamushi infection is still unknown. Using our recently developed BALB/c mouse model that is based on footpad inoculation of the human-pathogenic Karp strain, we show that activated CD8+ T cells infiltrate spleen and lung during the third week of infection. Depletion of CD8+ T cells with monoclonal antibodies resulted in uncontrolled pathogen growth and mortality. Adoptive transfer of CD8+ T cells from infected animals protected naïve BALB/c mice from lethal outcome of intraperitoneal challenge. In C57Bl/6 mice, the pulmonary lymphocyte compartment showed an increased percentage of CD8+ T cells for at least 135 days post O. tsutsugamushi infection. Depletion of CD8+ T cells at 84 days post infection caused reactivation of bacterial growth. In CD8+ T cell-deficient beta 2-microglobulin knockout mice, bacterial replication was uncontrolled, and all mice succumbed to the infection, despite higher serum IFN-γ levels and stronger macrophage responses in liver and lung. Moreover, we show that CD8+ T cells but not NKT cells were required for hepatocyte injury: elevated concentrations of serum alanine aminotransferase and infection-induced subcapsular necrotic liver lesions surrounded by macrophages were found in C57Bl/6 and CD1d-deficient mice, but not in beta 2-microglobulin knockout mice. In the lungs, peribronchial macrophage infiltrations also depended on CD8+ T cells. In summary, our results demonstrate that CD8+ T cells restrict growth of O. tsutsugamushi during acute and persistent infection, and are required to protect from lethal infections in BALB/c and C57BL/6 mice. However, they also elicit specific pathologic tissue lesions in liver and lung.
Collapse
Affiliation(s)
- Matthias Hauptmann
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Julia Kolbaum
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stefanie Lilla
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - David Wozniak
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Mohammad Gharaibeh
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Bernhard Fleischer
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian A. Keller
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
25
|
Smalley C, Bechelli J, Rockx-Brouwer D, Saito T, Azar SR, Ismail N, Walker DH, Fang R. Rickettsia australis Activates Inflammasome in Human and Murine Macrophages. PLoS One 2016; 11:e0157231. [PMID: 27362650 PMCID: PMC4928923 DOI: 10.1371/journal.pone.0157231] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
Rickettsiae actively escape from vacuoles and replicate free in the cytoplasm of host cells, where inflammasomes survey the invading pathogens. In the present study, we investigated the interactions of Rickettsia australis with the inflammasome in both mouse and human macrophages. R. australis induced a significant level of IL-1β secretion by human macrophages, which was significantly reduced upon treatment with an inhibitor of caspase-1 compared to untreated controls, suggesting caspase-1-dependent inflammasome activation. Rickettsia induced significant secretion of IL-1β and IL-18 in vitro by infected mouse bone marrow-derived macrophages (BMMs) as early as 8-12 h post infection (p.i.) in a dose-dependent manner. Secretion of these cytokines was accompanied by cleavage of caspase-1 and was completely abrogated in BMMs deficient in caspase-1/caspase-11 or apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), suggesting that R. australis activate the ASC-dependent inflammasome. Interestingly, in response to the same quantity of rickettsiae, NLRP3-/- BMMs significantly reduced the secretion level of IL-1β compared to wild type (WT) controls, suggesting that NLRP3 inflammasome contributes to cytosolic recognition of R. australis in vitro. Rickettsial load in spleen, but not liver and lung, of R. australis-infected NLRP3-/- mice was significantly greater compared to WT mice. These data suggest that NLRP3 inflammasome plays a role in host control of bacteria in vivo in a tissue-specific manner. Taken together, our data, for the first time, illustrate the activation of ASC-dependent inflammasome by R. australis in macrophages in which NLRP3 is involved.
Collapse
Affiliation(s)
- Claire Smalley
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Jeremy Bechelli
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Dedeke Rockx-Brouwer
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tais Saito
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Sasha R. Azar
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Nahed Ismail
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - David H. Walker
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Rong Fang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
26
|
Nonselective Persistence of a Rickettsia conorii Extrachromosomal Plasmid during Mammalian Infection. Infect Immun 2016; 84:790-7. [PMID: 26755154 DOI: 10.1128/iai.01205-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/28/2015] [Indexed: 01/26/2023] Open
Abstract
Scientific analysis of the genus Rickettsia is undergoing a rapid period of change with the emergence of viable genetic tools. The development of these tools for the mutagenesis of pathogenic bacteria will permit forward genetic analysis of Rickettsia pathogenesis. Despite these advances, uncertainty still remains regarding the use of plasmids to study these bacteria in in vivo mammalian models of infection, namely, the potential for virulence changes associated with the presence of extrachromosomal DNA and nonselective persistence of plasmids in mammalian models of infection. Here, we describe the transformation of Rickettsia conorii Malish 7 with the plasmid pRam18dRGA[AmTrCh]. Transformed R. conorii stably maintains this plasmid in infected cell cultures, expresses the encoded fluorescent proteins, and exhibits growth kinetics in cell culture similar to those of nontransformed R. conorii. Using a well-established murine model of fatal Mediterranean spotted fever, we demonstrate that R. conorii(pRam18dRGA[AmTrCh]) elicits the same fatal outcomes in animals as its untransformed counterpart and, importantly, maintains the plasmid throughout infection in the absence of selective antibiotic pressure. Interestingly, plasmid-transformed R. conorii was readily observed both in endothelial cells and within circulating leukocytes. Together, our data demonstrate that the presence of an extrachromosomal DNA element in a pathogenic rickettsial species does not affect either in vitro proliferation or in vivo infectivity in models of disease and that plasmids such as pRam18dRGA[AmTrCh] are valuable tools for the further genetic manipulation of pathogenic rickettsiae.
Collapse
|
27
|
Milhano N, Saito TB, Bechelli J, Fang R, Vilhena M, DE Sousa R, Walker DH. The role of Rhipicephalus sanguineus sensu lato saliva in the dissemination of Rickettsia conorii in C3H/HeJ mice. MEDICAL AND VETERINARY ENTOMOLOGY 2015; 29:225-229. [PMID: 26011701 DOI: 10.1111/mve.12118] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/24/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023]
Abstract
Animal models have been developed for the study of rickettsial pathogenesis. However, to understand what occurs during the natural route of rickettsial transmission via the tick bite, the role of tick saliva should be considered in these models. To address this, we analysed the role of tick saliva in the transmission of Rickettsia conorii (Rickettsiales: Rickettsiaceae) in a murine host by intradermally (i.d.) inoculating two groups of susceptible C3H/HeJ mice with this Rickettsia, and infesting one group with nymphal Rhipicephalus sanguineus sensu lato (Ixodida: Ixodidae) ticks. Quantification of bacterial loads and mRNA levels of interleukin-1β (IL-1β), IL-10 and NF-κB was performed in C3H/HeJ lung samples by real-time quantitative polymerase chain reaction (PCR) and real-time reverse transcriptase PCR, respectively. Lung histology was examined to evaluate the pathological manifestations of infection. No statistically significant difference in bacterial load in the lungs of mice was observed between these two groups; however, a statistically significant difference was observed in levels of IL-1β and NF-κB, both of which were higher in the group inoculated with rickettsiae but not infected with ticks. Lung histology in both groups of animals revealed infiltration of inflammatory cells. Overall, this study showed that i.d. inoculation of R. conorii caused infection in the lungs of C3H/HeJ mice and tick saliva inhibited proinflammatory effects.
Collapse
Affiliation(s)
- N Milhano
- Centre for the Study of Vectors and Infectious Diseases Dr Francisco Cambournac, National Institute of Health Dr Ricardo Jorge, Águas de Moura, Portugal
| | - T B Saito
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - J Bechelli
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - R Fang
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - M Vilhena
- Department of Veterinary Medicine, University of Évora, Évora, Portugal
| | - R DE Sousa
- Centre for the Study of Vectors and Infectious Diseases Dr Francisco Cambournac, National Institute of Health Dr Ricardo Jorge, Águas de Moura, Portugal
| | - D H Walker
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| |
Collapse
|
28
|
Increased Nucleosomes and Neutrophil Activation Link to Disease Progression in Patients with Scrub Typhus but Not Murine Typhus in Laos. PLoS Negl Trop Dis 2015; 9:e0003990. [PMID: 26317419 PMCID: PMC4552835 DOI: 10.1371/journal.pntd.0003990] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 07/15/2015] [Indexed: 11/19/2022] Open
Abstract
Cell-mediated immunity is essential in protection against rickettsial illnesses, but the role of neutrophils in these intracellular vasculotropic infections remains unclear. This study analyzed the plasma levels of nucleosomes, FSAP-activation (nucleosome-releasing factor), and neutrophil activation, as evidenced by neutrophil-elastase (ELA) complexes, in sympatric Lao patients with scrub typhus and murine typhus. In acute scrub typhus elevated nucleosome levels correlated with lower GCS scores, raised respiratory rate, jaundice and impaired liver function, whereas neutrophil activation correlated with fibrinolysis and high IL-8 plasma levels, a recently identified predictor of severe disease and mortality. Nucleosome and ELA complex levels were associated with a 4.8-fold and 4-fold increased risk of developing severe scrub typhus, beyond cut off values of 1,040 U/ml for nucleosomes and 275 U/ml for ELA complexes respectively. In murine typhus, nucleosome levels associated with pro-inflammatory cytokines and the duration of illness, while ELA complexes correlated strongly with inflammation markers, jaundice and increased respiratory rates. This study found strong correlations between circulating nucleosomes and neutrophil activation in patients with scrub typhus, but not murine typhus, providing indirect evidence that nucleosomes could originate from neutrophil extracellular trap (NET) degradation. High circulating plasma nucleosomes and ELA complexes represent independent risk factors for developing severe complications in scrub typhus. As nucleosomes and histones exposed on NETs are highly cytotoxic to endothelial cells and are strongly pro-coagulant, neutrophil-derived nucleosomes could contribute to vascular damage, the pro-coagulant state and exacerbation of disease in scrub typhus, thus indicating a detrimental role of neutrophil activation. The data suggest that increased neutrophil activation relates to disease progression and severe complications, and increased plasma levels of nucleosomes and ELA complexes represent independent risk factors for developing severe scrub typhus. Tropical rickettsial illnesses, especially scrub typhus and murine typhus, are increasingly recognized as a leading cause of treatable undifferentiated febrile illness in Asia, but remain severely neglected and under appreciated diseases in many areas. In this study we investigated the relationship of markers of neutrophil activation and cell death with disease severity in patients with acute scrub typhus and murine typhus in Laos. These easily measurable circulating markers were associated with a 4 to 5-fold increased risk of developing severe clinical disease manifestations in scrub typhus and represent independent predictors of severe disease, and possibly death. We also found strong correlations between circulating markers of cell death and neutrophil activation in patients with scrub typhus, but not murine typhus, providing indirect evidence that neutrophil extracellular traps could contribute to the vascular damage and pro-coagulant state leading to exacerbation of disease in scrub typhus, thus indicating a detrimental role of neutrophil activation. The data suggest that increased neutrophil activation relates to disease progression and severe complications, and increased plasma levels of nucleosomes and ELA complexes represent independent risk factors for developing severe scrub typhus.
Collapse
|
29
|
Soong L, Wang H, Shelite TR, Liang Y, Mendell NL, Sun J, Gong B, Valbuena GA, Bouyer DH, Walker DH. Strong type 1, but impaired type 2, immune responses contribute to Orientia tsutsugamushi-induced pathology in mice. PLoS Negl Trop Dis 2014; 8:e3191. [PMID: 25254971 PMCID: PMC4177881 DOI: 10.1371/journal.pntd.0003191] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022] Open
Abstract
Scrub typhus is a neglected, but important, tropical disease, which puts one-third of the world's population at risk. The disease is caused by Orientia tsutsugamushi, an obligately intracellular Gram-negative bacterium. Dysregulation in immune responses is known to contribute to disease pathogenesis; however, the nature and molecular basis of immune alterations are poorly defined. This study made use of a newly developed murine model of severe scrub typhus and focused on innate regulators and vascular growth factors in O. tsutsugamushi-infected liver, lungs and spleen. We found no activation or even reduction in base-line expression for multiple molecules (IL-7, IL-4, IL-13, GATA3, ROR-γt, and CXCL12) at 2, 6 and 10 days post-infection. This selective impairment in type 2-related immune responses correlated with a significant activation of the genes for IL-1β, IL-6, IL-10, TNF-α, IFN-γ, as well as CXCR3- and CXCR1-related chemokines in inflamed tissues. The elevated angiopoietin (Ang)-2 expression and Ang-2/Ang-1 ratios suggested excessive inflammation and the loss of endothelial integrity. These alterations, together with extensive recruitment of myeloperoxidase (MPO)-expressing neutrophils and the influx of CD3+ T cells, contributed to acute tissue damage and animal death. This is the first report of selective alterations in a panel of immune regulators during early O. tsutsugamushi infection in intravenously inoculated C57BL/6 mice. Our findings shed new light on the pathogenic mechanisms associated with severe scrub typhus and suggest potential targets for therapeutic investigation.
Collapse
Affiliation(s)
- Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| | - Hui Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas R. Shelite
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nicole L. Mendell
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Gustavo A. Valbuena
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H. Bouyer
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David H. Walker
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
30
|
Phenotype of the anti-Rickettsia CD8(+) T cell response suggests cellular correlates of protection for the assessment of novel antigens. Vaccine 2014; 32:4960-7. [PMID: 25043277 DOI: 10.1016/j.vaccine.2014.07.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 06/19/2014] [Accepted: 07/08/2014] [Indexed: 02/03/2023]
Abstract
The obligately intracellular bacteria Rickettsia infect endothelial cells and cause systemic febrile diseases that are potentially lethal. No vaccines are currently available and current knowledge of the effective immune response is limited. Natural and experimental rickettsial infections provide strong and cross-protective cellular immunity if the infected individual survives the acute infection. Although resistance to rickettsial infections is attributed to the induction of antigen-specific T cells, particularly CD8(+) T cells, the identification and validation of correlates of protective cellular immunity against rickettsial infections, an important step toward vaccine validation, remains a gap in this field. Here, we show that after a primary challenge with Rickettsia typhi in the C3H mouse model, the peak of anti-Rickettsia CD8(+) T cell-mediated responses occurs 7 days post-infection (dpi), which coincides with the beginning of rickettsial clearance. At this time point, both effector-type and memory-type CD8(+) T cells are present, suggesting that 7 dpi is a valid time point for the assessment of CD8(+) T cell responses of mice previously immunized with protective antigens. Based on our results, we suggest four correlates of cellular protection for the assessment of protective rickettsial antigens: (1) production of IFN-γ by antigen-experienced CD3(+)CD8(+)CD44(high) cells, (2) production of Granzyme B by CD27(low)CD43(low) antigen-experienced CD8(+) T cells, (3) generation of memory-type CD8(+) T cells [Memory Precursor Effector Cells (MPECs), as well as CD127(high)CD43(low), and CD27(high)CD43(low) CD8(+) T cells], and (4) generation of effector-like memory CD8(+) T cells (CD27(low)CD43(low)). We propose that these correlates could be useful for the general assessment of the quality of the CD8(+) T cell immune response induced by novel antigens with potential use in a vaccine against Rickettsia.
Collapse
|
31
|
Shelite TR, Saito TB, Mendell NL, Gong B, Xu G, Soong L, Valbuena G, Bouyer DH, Walker DH. Hematogenously disseminated Orientia tsutsugamushi-infected murine model of scrub typhus [corrected]. PLoS Negl Trop Dis 2014; 8:e2966. [PMID: 25010338 PMCID: PMC4091938 DOI: 10.1371/journal.pntd.0002966] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/24/2014] [Indexed: 11/18/2022] Open
Abstract
Orientia tsutsugamushi, the etiologic agent of scrub typhus, is a mite-borne rickettsia transmitted by the parasitic larval stage of trombiculid mites. Approximately one-third of the world's population is at risk of infection with Orientia tsutsugamushi, emphasizing its importance in global health. In order to study scrub typhus, Orientia tsutsugamushi Karp strain has been used extensively in mouse studies with various inoculation strategies and little success in inducing disease progression similar to that of human scrub typhus. The objective of this project was to develop a disease model with pathology and target cells similar to those of severe human scrub typhus. This study reports an intravenous infection model of scrub typhus in C57BL/6 mice. This mouse strain was susceptible to intravenous challenge, and lethal infection occurred after intravenous inoculation of 1.25×106 focus (FFU) forming units. Signs of illness in lethally infected mice appeared on day 6 with death occurring ∼6 days later. Immunohistochemical staining for Orientia antigens demonstrated extensive endothelial infection, most notably in the lungs and brain. Histopathological analysis revealed cerebral perivascular, lymphohistiocytic infiltrates, focal hemorrhages, meningoencephalitis, and interstitial pneumonia. Disseminated infection of endothelial cells with Orientia in C57BL/6 mice resulted in pathology resembling that of human scrub typhus. The use of this model will allow detailed characterization of the mechanisms of immunity to and pathogenesis of O. tsutsugamushi infection. Scrub typhus is a disease found in Southeast Asia that infects over 1 million people each year. This disease is caused by the intracellular pathogen Orientia tsutsugamushi transmitted by the bite of chigger mites. Scrub typhus is characterized by pulmonary disease and in severe cases, multiorgan system failure. The current research model utilizes an intraperitoneal route of inoculation of mice to study the host response to Orientia infection. Infection via this route results in severe peritonitis that does not occur in human scrub typhus. The development of animal models that accurately portray human disease is an important step toward understanding and managing disease. In this manuscript we describe a new mouse model that results in scrub typhus-like pathology following intravenous inoculation of mice. This model presents dose-dependent mortality with scrub typhus-like pathology that parallels human disease. Utilization of this model will provide a valuable research tool for characterizing the immune response and pathogenesis induced by O. tsutsugamushi allowing development of better treatment and an effective vaccine.
Collapse
Affiliation(s)
- Thomas R. Shelite
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Tais B. Saito
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nicole L. Mendell
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Guang Xu
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Gustavo Valbuena
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H. Bouyer
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David H. Walker
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Caro-Gomez E, Gazi M, Goez Y, Valbuena G. Discovery of novel cross-protective Rickettsia prowazekii T-cell antigens using a combined reverse vaccinology and in vivo screening approach. Vaccine 2014; 32:4968-76. [PMID: 25010827 DOI: 10.1016/j.vaccine.2014.06.089] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 06/04/2014] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
Abstract
Rickettsial agents are some of the most lethal pathogens known to man. Among them, Rickettsia prowazekii is a select agent with potential use for bioterrorism; yet, there is no anti-Rickettsia vaccine commercially available. Owing to the obligate intracellular lifestyle of rickettsiae, CD8(+) T cells are indispensable for protective cellular immunity. Furthermore, T cells can mediate cross-protective immunity between different pathogenic Rickettsia, a finding consistent with the remarkable similarity among rickettsial genomes. However, Rickettsia T cell antigens remain unidentified. In the present study, we report an algorithm that allowed us to identify and validate four novel R. prowazekii vaccine antigen candidates recognized by CD8(+) T cells from a set of twelve in silico-defined protein targets. Our results highlight the importance of combining proteasome-processing as well as MHC class-I-binding predictions. The novel rickettsial vaccine candidate antigens, RP778, RP739, RP598, and RP403, protected mice against a lethal challenge with Rickettsia typhi, which is indicative of cross-protective immunity within the typhus group rickettsiae. Together, our findings validate a reverse vaccinology approach as a viable strategy to identify protective rickettsial antigens and highlight the feasibility of a subunit vaccine that triggers T-cell-mediated cross-protection among diverse rickettsiae.
Collapse
Affiliation(s)
- Erika Caro-Gomez
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA
| | - Michal Gazi
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA
| | - Yenny Goez
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA
| | - Gustavo Valbuena
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609, USA; Sealy Center for Vaccine Development, Center for Tropical Diseases, Center for Biodefense and Emerging Infectious Diseases, Institute for Translational Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| |
Collapse
|
33
|
Sahni SK, Narra HP, Sahni A, Walker DH. Recent molecular insights into rickettsial pathogenesis and immunity. Future Microbiol 2014; 8:1265-88. [PMID: 24059918 DOI: 10.2217/fmb.13.102] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human infections with arthropod-borne Rickettsia species remain a major global health issue, causing significant morbidity and mortality. Epidemic typhus due to Rickettsia prowazekii has an established reputation as the 'scourge of armies', and as a major determinant of significant 'historical turning points'. No suitable vaccines for human use are currently available to prevent rickettsial diseases. The unique lifestyle features of rickettsiae include obligate intracellular parasitism, intracytoplasmic niche within the host cell, predilection for infection of microvascular endothelium in mammalian hosts, association with arthropods and the tendency for genomic reduction. The fundamental research in the field of Rickettsiology has witnessed significant recent progress in the areas of pathogen adhesion/invasion and host immune responses, as well as the genomics, proteomics, metabolomics, phylogenetics, motility and molecular manipulation of important rickettsial pathogens. The focus of this review article is to capture a snapshot of the latest developments pertaining to the mechanisms of rickettsial pathogenesis and immunity.
Collapse
Affiliation(s)
- Sanjeev K Sahni
- Department of Pathology & Institute for Human Infections & Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | | | | | | |
Collapse
|
34
|
Discovery of a protective Rickettsia prowazekii antigen recognized by CD8+ T cells, RP884, using an in vivo screening platform. PLoS One 2013; 8:e76253. [PMID: 24146844 PMCID: PMC3797808 DOI: 10.1371/journal.pone.0076253] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/22/2013] [Indexed: 01/22/2023] Open
Abstract
Rickettsia prowazekii has been tested for biological warfare due to the high mortality that it produces after aerosol transmission of very low numbers of rickettsiae. Epidemic typhus, the infection caused by these obligately intracellular bacteria, continues to be a threat because it is difficult to diagnose due to initial non-specific symptoms and the lack of commercial diagnostic tests that are sensitive and specific during the initial clinical presentation. A vaccine to prevent epidemic typhus would constitute an effective deterrent to the weaponization of R. prowazekii; however, an effective and safe vaccine is not currently available. Due to the cytoplasmic niche of Rickettsia, CD8+ T-cells are critical effectors of immunity; however, the identification of antigens recognized by these cells has not been systematically addressed. To help close this gap, we designed an antigen discovery strategy that uses cell-based vaccination with antigen presenting cells expressing microbe's proteins targeted to the MHC class I presentation pathway. We report the use of this method to discover a protective T-cell rickettsial antigen, RP884, among a test subset of rickettsial proteins.
Collapse
|
35
|
Fang R, Ismail N, Walker DH. Contribution of NK cells to the innate phase of host protection against an intracellular bacterium targeting systemic endothelium. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:185-95. [PMID: 22617213 DOI: 10.1016/j.ajpath.2012.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 02/10/2012] [Accepted: 03/22/2012] [Indexed: 11/19/2022]
Abstract
We investigated the mechanisms by which natural killer (NK) cells mediate innate host defense against infection with an endothelium-targeting intracellular bacterium, Rickettsia. We found that a robust Rickettsia-induced innate response in resistant mice cleared the bacteria early in the infection and was associated with significantly higher frequencies of splenic interferon (IFN)-γ (+) CD8(+) T cells and cytotoxic NK cells compared with susceptible mice. More importantly, NK cell-deficient Rag(-/-)γc(-/-) animals displayed significantly increased susceptibility to Rickettsia infection compared with NK cell-sufficient Rag(-/-) mice, as evidenced by impaired bacterial clearance, early development of severe thrombosis in the liver, and a decreased serum level of IFN-γ. Furthermore, the lack of NK cells also impaired host resistance of CB-17 scid mice to Rickettsia, similar to what was observed in Rag(-/-)γc(-/-) mice. Interestingly, perforin deficiency in Rag(-/-)Prf1(-/-) mice resulted in greater thrombosis and insignificantly different systemic levels of IFN-γ compared with Rag(-/-) mice, suggesting that perforin, which is mainly produced by NK cells, is involved in the prevention of vascular damage. Together, these findings reveal that NK cells mediate the innate phase of host protection against infection with rickettsiae, most likely via IFN-γ production. Furthermore, NK cells are involved in preventing rickettsial infection-induced endothelial cell damage, possibly via perforin production.
Collapse
Affiliation(s)
- Rong Fang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
| | | | | |
Collapse
|
36
|
Abstract
Rickettsia parkeri, a member of the spotted fever group Rickettsia, is the causative agent of American boutonneuse fever in humans. Despite the increased recognition of human cases, limited information is available regarding the infection of invertebrate and vertebrate hosts for this emerging tick-borne disease. Toward the development of a viable transmission model and to further characterize the pathology associated with R. parkeri infection, inbred mouse strains (A/J, BALB/c, C3H/HeJ, and C3H/HeN) were intravenously and intradermally inoculated with 10(5) low-passage-number R. parkeri (Portsmouth strain), and infection, gross pathology, and histopathology were scored. Additionally, a quantitative real-time PCR (qPCR) was performed to estimate rickettsial load in heart, lung, spleen, and liver tissues of infected mice at 19 days postinoculation. Of the A/J, BALB/c, and C3H/HeN mice, none displayed universal pathology consistent with sustained infection. Compared to age-matched control mice, the intravenously inoculated C3H/HeJ mice exhibited marked facial edema and marked splenomegaly upon gross examination, while the intradermally inoculated mice developed characteristic eschar-like lesions. The C3H/HeJ mice also exhibited the greatest concentrations of rickettsial DNA from heart, lung, liver, and spleen samples when examined by qPCR. The similarity of the pathology of human disease and sustained infection suggests that the C3H/HeJ strain of mice is a promising candidate for subsequent experiments to examine the tick transmission, dissemination, and pathology of R. parkeri rickettsiosis.
Collapse
|
37
|
Paris DH, Chansamouth V, Nawtaisong P, Löwenberg EC, Phetsouvanh R, Blacksell SD, Lee SJ, Dondorp AM, van der Poll T, Newton PN, Levi M, Day NPJ. Coagulation and inflammation in scrub typhus and murine typhus--a prospective comparative study from Laos. Clin Microbiol Infect 2011; 18:1221-8. [PMID: 22192733 PMCID: PMC3533763 DOI: 10.1111/j.1469-0691.2011.03717.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Scrub typhus (caused by Orientia tsutsugamushi) and murine typhus (caused by Rickettsia typhi) cause up to 28% of febrile episodes in Thailand and Laos. The current understanding of coagulation and inflammation in the pathogenesis of these clinically very similar vasculotropic diseases is limited. This study compared human in vivo changes in 15 coagulation, inflammation and endothelial activation markers in prospectively collected admission and follow-up samples of 121 patients (55 scrub typhus, 55 murine typhus, and 11 typhus-like illness) and 51 healthy controls from Laos. As compared with controls, all but one of the markers assessed were significantly affected in typhus patients; however, the activation patterns differed significantly between scrub and murine typhus patients. The levels of markers of coagulation activation and all inflammatory cytokines, except for interleukin-12, were significantly higher in patients with scrub typhus than in those with murine typhus. In patients with murine typhus, however, the levels of endothelium-derived markers were significantly higher. Anticoagulant factors were inhibited in both typhus patient groups. This is the first study demonstrating that, in scrub typhus, in vivo coagulation activation is prominent and is related to a strong proinflammatory response, whereas in murine typhus, changes in coagulant and fibrinolytic pathways are suggestive of endothelial cell perturbation. These data suggest that, although late-stage endothelial infection is common in both diseases, the in vivo pathogenic mechanisms of R. typhi and O. tsutsugamushi could differ in the early phase of infection and may contribute to disease differentiation.
Collapse
Affiliation(s)
- D H Paris
- Mahidol-Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Beta interferon-mediated activation of signal transducer and activator of transcription protein 1 interferes with Rickettsia conorii replication in human endothelial cells. Infect Immun 2011; 79:3733-43. [PMID: 21690236 DOI: 10.1128/iai.05008-11] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Infection of the endothelial cell lining of blood vessels with Rickettsia conorii, the causative agent of Mediterranean spotted fever, results in endothelial activation. We investigated the effects of R. conorii infection on the status of the Janus kinase (JAK)-signal transducer and activator of transcription protein (STAT) signaling pathway in human microvascular endothelial cells (HMECs), the most relevant host cell type, in light of rickettsial tropism for microvascular endothelium in vivo. R. conorii infection induced phosphorylation of STAT1 on tyrosine 701 and serine 727 at 24, 48, and 72 h postinfection in HMECs. Employing transcription profile analysis and neutralizing antibodies, we further determined that beta interferon (IFN-β) production and secretion are critical for STAT1 activation. Secreted IFN-β further amplified its own expression via a positive-feedback mechanism, while expression of transcription factors interferon regulatory factor 7 (IRF7) and IRF9, implicated in the IFN-β-STAT1 feedback loop, was also induced. Metabolic activity of rickettsiae was essential for the IFN-β-mediated response(s) because tetracycline treatment inhibited R. conorii replication, IFN-β expression, and STAT1 phosphorylation. Inclusion of IFN-β-neutralizing antibody during infection resulted in significantly enhanced R. conorii replication, whereas addition of exogenous IFN-β had the opposite inhibitory effect. Finally, small interfering RNA-mediated knockdown further confirmed a protective role for STAT1 against intracellular R. conorii replication. In concert, these findings implicate an important role for IFN-β-mediated STAT1 activation in innate immune responses of vascular endothelium to R. conorii infection.
Collapse
|
39
|
Walker DH. The realities of biodefense vaccines against Rickettsia. Vaccine 2009; 27 Suppl 4:D52-5. [PMID: 19837287 PMCID: PMC2909128 DOI: 10.1016/j.vaccine.2009.07.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 07/15/2009] [Indexed: 11/22/2022]
Abstract
Rickettsia prowazekii, R. rickettsii, R. conorii, and R. typhi are serious biologic weapon threats because of high infectivity of low dose aerosols, stable small particle aerosol infectivity, virulence causing severe disease, difficulty in establishing a timely diagnosis, ineffectiveness of usual empiric treatments, potential for engineered complete antimicrobial resistance, lower level of immunity, availability of the agents in nature, and feasibility of propagation, stabilization, and dispersal. Infection induces long-term immunity, killed rickettsial vaccines stimulate incomplete protection, and a live attenuated mutant stimulates strong immunity but reverts to virulence. Prospects for rational development of a safe, effective live attenuated vaccine are excellent.
Collapse
Affiliation(s)
- David H Walker
- University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0609, USA.
| |
Collapse
|
40
|
Sahni SK, Rydkina E. Host-cell interactions with pathogenic Rickettsia species. Future Microbiol 2009; 4:323-39. [PMID: 19327117 DOI: 10.2217/fmb.09.6] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Sanjeev K Sahni
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | |
Collapse
|
41
|
Bechah Y, Capo C, Mege JL, Raoult D. Rickettsial diseases: from Rickettsia-arthropod relationships to pathophysiology and animal models. Future Microbiol 2008; 3:223-36. [PMID: 18366341 DOI: 10.2217/17460913.3.2.223] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rickettsiae cause spotted fevers and typhus-related diseases in humans. Some of these diseases occur worldwide and are life-threatening, for example, epidemic typhus is still a major health problem despite the apparent efficiency of antibiotic treatment. In addition, Rickettsia prowazekii, the agent of epidemic typhus, and R. rickettsii, the agent of Rocky Mountain spotted fever, are microorganisms that could potentially be used as bioweapons to induce panic in the population. Rickettsiae are obligate intracellular bacteria in both vertebrate and invertebrate hosts, but rickettsial species differ in terms of association with arthropods, behavior of the vector to infection, pathophysiology and outcome of the disease. Understanding the pathogenic steps of rickettsioses is essential to develop protective strategies against these bacteriological threats. Unfortunately, the mechanisms involved in the pathogenesis of many rickettsioses are poorly characterized, and protective immunity is incompletely understood, in part because accurate animal models that mimic human diseases are lacking. In the past, murine models have been of limited value because infection of mice was without effect or resulted in erratic mortality. Recent studies have reported that rickettsial infection can be established in mice, depending on the genetic background of mice, the type of rickettsial species and the route of inoculation. These models may be useful for analyzing the pathogenesis of rickettsioses, especially epidemic typhus, evaluating new therapeutic molecules and vaccine candidates, and preventing future outbreaks.
Collapse
Affiliation(s)
- Yassina Bechah
- Unité des Rickettsies, CNRS UMR 6020, Université de la Méditerranée, Faculté de Médecine, 27 Bld. Jean Moulin, 13385 Marseille Cedex 05, France.
| | | | | | | |
Collapse
|
42
|
Valbuena G, Walker DH. The endothelium as a target for infections. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2007; 1:171-98. [PMID: 18039112 DOI: 10.1146/annurev.pathol.1.110304.100031] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endothelial cells lining vascular and lymphatic vessels are targets of several infectious agents, including viruses and bacteria, that lead to dramatic changes in their functions. Understanding the pathophysiological mechanisms that cause the clinical manifestations of those infections has been advanced through the use of animal models and in vitro systems; however, there are also abundant studies that explore the consequences of endothelial infection in vitro without supporting evidence that endothelial cells are actual in vivo targets of infection in human diseases. This article defines criteria for considering an infection as truly endothelium-targeted and reviews the literature that offers insights into the pathogenesis of human endothelial-target infections.
Collapse
|
43
|
Woods ME, Olano JP. Host defenses to Rickettsia rickettsii infection contribute to increased microvascular permeability in human cerebral endothelial cells. J Clin Immunol 2007; 28:174-85. [PMID: 17957455 DOI: 10.1007/s10875-007-9140-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Accepted: 09/19/2007] [Indexed: 12/07/2022]
Abstract
Rickettsiae are arthropod-borne intracellular bacterial pathogens that primarily infect the microvascular endothelium leading to systemic spread of the organisms and the major pathophysiological effect, increased microvascular permeability, and edema in vital organs such as the lung and brain. Much work has been done on mechanisms of immunity to rickettsiae, as well as the responses of endothelial cells to rickettsial invasion. However, to date, no one has described the mechanisms of increased microvascular permeability during acute rickettsiosis. We sought to establish an in vitro model of human endothelial-target rickettsial infection using the etiological agent of Rocky Mountain spotted fever, Rickettsia rickettsii, and human cerebral microvascular endothelial cells. Endothelial cells infected with R. rickettsii exhibited a dose-dependent decrease in trans-endothelial electrical resistance, which translates into increased monolayer permeability. Additionally, we showed that the addition of pro-inflammatory stimuli essential to rickettsial immunity dramatically enhanced this effect. This increase in permeability correlates with dissociation of adherens junctions between endothelial cells and is not dependent on the presence of nitric oxide. Taken together, these results demonstrate for the first time that increased microvascular permeability associated with rickettsial infection is partly attributable to intracellular rickettsiae and partly attributable to the immune defenses that have evolved to protect the host from rickettsial spread.
Collapse
Affiliation(s)
- Michael E Woods
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd. Rt 0428, Galveston, TX, USA
| | | |
Collapse
|
44
|
Svraka S, Rolain JM, Bechah Y, Gatabazi J, Raoult D. Rickettsia prowazekii and real-time polymerase chain reaction. Emerg Infect Dis 2006; 12:428-32. [PMID: 16704780 PMCID: PMC3291444 DOI: 10.3201/eid1203.050888] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
This highly standardized and adaptable assay could improve epidemic typhus surveillance. Rickettsia prowazekii is the causative agent of epidemic typhus and a potential bioterrorism agent. Sensitive and specific rapid assays are needed to complement existing methods of detecting this organism. We developed a real-time quantitative polymerase chain reaction assay by using a species-specific probe targeting the gltA gene. This assay, which was rapid, specific for R. prowazekii only, and sensitive (cutoff detection of 1 to 5 copies per sample), detected and directly identified R. prowazekii in blood of 12 experimentally infected mice sampled at day 3 and 6 postinfection or in naturally or experimentally infected lice. Because our assay is highly standardized and easily adaptable, it could improve epidemic typhus surveillance in public health programs, especially for countries with underdiagnosed or unrecognized human cases.
Collapse
|
45
|
Abstract
Rickettsiae are obligate intracellular alpha-proteobacteria that primarily target the microvascular endothelium. In the last two decades, new rickettsial pathogens have been associated with human illness around the world. Clinically, the common denominator in all rickettsioses is the development of increased microvascular permeability, leading to cerebral and non-cardiogenic pulmonary edema. With the development of powerful research tools, advances in the understanding of rickettsial pathogenesis have been dramatic. Entry into the host cell is followed by rapid escape into the cytoplasm to avoid phagolysosomal fusion. Spotted fever group rickettsiae induce actin polymerization via a group of proteins called RickA, which promote nucleation of actin monomers via the Arp2/3 complex at one rickettsial pole, propelling the bacteria across the cytoplasm and into neighboring cells. Damage to the host cell is most likely multifactorial. The most extensively studied mechanism is the generation of reactive oxygen species (ROS) and downregulation of enzymes involved in protection against oxidative injury. The significance of ROS-mediated cellular damage in vivo is beginning to be elucidated. The main pathogenic mechanism is increased microvascular permeability leading to profound metabolic disturbances in the extravascular compartment. The underlying factors responsible for those changes are beginning to be elucidated in vitro and include direct effects of intracellular rickettsiae, cytokines, and possibly activated coagulation factors--all of which most likely modify interendothelial junctions. Our knowledge on rickettsial pathogenesis will continue to expand in the near future as new research tools become available.
Collapse
Affiliation(s)
- Juan P Olano
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0609, USA.
| |
Collapse
|
46
|
Loftis AD, Nicholson WL, Levin ML. Evaluation of immunocompetent and immunocompromised mice (Mus musculus) for infection with Ehrlichia chaffeensis and transmission to Amblyomma americanum ticks. Vector Borne Zoonotic Dis 2005; 4:323-33. [PMID: 15671738 DOI: 10.1089/vbz.2004.4.323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Experiments on the natural history of Ehrlichia chaffeensis, the agent of human monocytic ehrlichiosis (HME), would be facilitated by the availability of a laboratory animal model for transmission to vector ticks. Five strains of mice were evaluated for their susceptibility to infection with E. chaffeensis and transmission competence: C57BL/6 mice, inducible nitric oxide synthase (iNOS) deficient mice, MHC I deficient (beta2m /) mice, MHC II deficient mice (Abb /), and B and T cell deficient (Rag1 /) mice. Mice were inoculated with a low passage isolate of E. chaffeensis, and infection and morbidity were monitored for 57 days. Three xenodiagnostic infestations with A. americanum nymphs were performed 1, 8, and 15 days following inoculation. C57BL/6 mice cleared the organism in less than 17 days, with no indication of morbidity, and mounted a rapid, strong antibody response. Transmission to feeding A. americanum nymphs was seen in 1/30 nymphs fed on C57BL/6 mice immediately after inoculation. In MHC I and iNOS deficient mice, pathogen DNA was detected up to 17 or 24 days, respectively, after inoculation. Persistent infection for the duration of the experiment (57 days) was observed in MHC II deficient mice. However, E. chaffeensis was not detected in ticks fed on iNOS, MHC I, or MHC II deficient mice. Susceptibility to infection was greatest in Rag1 knockout mice, with significant morbidity and mortality within 24 days after inoculation. E. chaffeensis DNA was detected in up to 55% of replete nymphs that fed on Rag1 mice. However, E. chaffeensis was not detected in molted adult ticks from the same cohorts.
Collapse
Affiliation(s)
- Amanda D Loftis
- Viral and Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA.
| | | | | |
Collapse
|
47
|
Valbuena G, Walker DH. Expression of CX3CL1 (fractalkine) in mice with endothelial-target rickettsial infection of the spotted-fever group. Virchows Arch 2004; 446:21-7. [PMID: 15480764 DOI: 10.1007/s00428-004-1120-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 08/19/2004] [Indexed: 12/16/2022]
Abstract
Fractalkine (CX3CL1) is a chemokine expressed mainly by endothelial cells, which are the major cellular targets of rickettsiae. We used immunohistochemistry to investigate the normal expression of CX3CL1 in mice and the kinetics of expression of this chemokine throughout the course of lethal and sublethal rickettsial infections in a mouse model of spotted-fever group rickettsioses. The peak of expression of fractalkine on day 3 of infection coincided with the time of infiltration of macrophages into infected tissues and preceded the peak of rickettsial content in tissues.
Collapse
Affiliation(s)
- Gustavo Valbuena
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-0609, USA
| | | |
Collapse
|
48
|
Wang X, Kang H, Kikuchi T, Suzuki Y. Gamma interferon production, but not perforin-mediated cytolytic activity, of T cells is required for prevention of toxoplasmic encephalitis in BALB/c mice genetically resistant to the disease. Infect Immun 2004; 72:4432-8. [PMID: 15271900 PMCID: PMC470633 DOI: 10.1128/iai.72.8.4432-4438.2004] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously showed the requirement of both T cells and gamma interferon (IFN-gamma)-producing non-T cells for the genetic resistance of BALB/c mice to the development of toxoplasmic encephalitis (TE). In order to define the role of IFN-gamma production and the perforin-mediated cytotoxicity of T cells in this resistance, we obtained immune T cells from spleens of infected IFN-gamma knockout (IFN-gamma(-/-)), perforin knockout (PO), and wild-type BALB/c mice and transferred them into infected and sulfadiazine-treated athymic nude mice, which lack T cells but have IFN-gamma-producing non-T cells. Control nude mice that had not received any T cells developed severe TE and died after discontinuation of sulfadiazine treatment due to the reactivation of infection. Animals that had received immune T cells from either wild-type or PO mice did not develop TE and survived. In contrast, nude mice that had received immune T cells from IFN-gamma(-/-) mice developed severe TE and died as early as control nude mice. T cells obtained from the spleens of animals that had received either PO or wild-type T cells produced large amounts of IFN-gamma after stimulation with Toxoplasma gondii antigens in vitro. In addition, the amounts of IFN-gamma mRNA expressed in the brains of PO T-cell recipients did not differ from those in wild-type T-cell recipients. Furthermore, PO mice did not develop TE after infection, and their IFN-gamma production was equivalent to or higher than that of wild-type animals. These results indicate that IFN-gamma production, but not perforin-mediated cytotoxic activity, by T cells is required for the prevention of TE in genetically resistant BALB/c mice.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Brain/parasitology
- Encephalitis/genetics
- Encephalitis/immunology
- Encephalitis/parasitology
- Female
- Humans
- Immunity, Innate/genetics
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Perforin
- Pore Forming Cytotoxic Proteins
- T-Lymphocytes/immunology
- Toxoplasma/pathogenicity
- Toxoplasmosis, Animal/genetics
- Toxoplasmosis, Animal/immunology
- Toxoplasmosis, Animal/mortality
- Toxoplasmosis, Animal/parasitology
- Toxoplasmosis, Cerebral/genetics
- Toxoplasmosis, Cerebral/immunology
- Toxoplasmosis, Cerebral/mortality
- Toxoplasmosis, Cerebral/parasitology
Collapse
Affiliation(s)
- Xisheng Wang
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | | | |
Collapse
|
49
|
Feng HM, Whitworth T, Olano JP, Popov VL, Walker DH. Fc-dependent polyclonal antibodies and antibodies to outer membrane proteins A and B, but not to lipopolysaccharide, protect SCID mice against fatal Rickettsia conorii infection. Infect Immun 2004; 72:2222-8. [PMID: 15039346 PMCID: PMC375156 DOI: 10.1128/iai.72.4.2222-2228.2004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An emphasis on cellular immunity against Rickettsia has led to neglect of analysis of the role of antibody. The availability of an excellent mouse model of spotted fever rickettsiosis enabled investigation of a potential role of antibody in immunity to Rickettsia conorii. C3H severe combined immunodeficiency (SCID) mice were passively transfused with monoclonal antibodies against rickettsial outer membrane protein A (OmpA), OmpB, or lipopolysaccharide (LPS), polyclonal anti-R. conorii serum, Fab fragments of polyclonal antiserum, or no antibodies and then challenged 48 h later with 10 50% lethal doses (LD(50)) of R. conorii. All mice that received monoclonal antibodies against OmpA and two of four mice that received monoclonal antibodies against OmpB or polyclonal antisera were completely protected, but the recipients of anti-LPS antibodies or the Fab fragments were not protected. Polyclonal antibody treatment of C3H SCID mice that had been infected with 10 LD(50) of R. conorii 4 or 5 days earlier prolonged the life of the infected mice from 10.4 to 22.5 days and resulted in decreased levels of infectious rickettsiae in the spleen and liver 24 and 48 h later. Treatment with protective antibodies resulted in the development of large aggregates of R. conorii antigens in splenic macrophages and intraphagolysosomal rickettsial death and digestion. The kinetics of development of antibodies to R. conorii determined by immunoblotting revealed antibodies to LPS on day 6 and antibodies to OmpA and OmpB on day 12, when recovery from the infection had already occurred. Antibodies to particular epitopes of OmpA and OmpB may protect against reinfection, but they may not play a key role in immunity against primary infection. Antibodies might be useful for treating infections with antibiotic-resistant organisms, and some B-cell epitopes should be included in a subunit vaccine.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/administration & dosage
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/immunology
- Bacterial Outer Membrane Proteins/immunology
- Boutonneuse Fever/mortality
- Boutonneuse Fever/prevention & control
- Immunization, Passive
- Lipopolysaccharides/immunology
- Mice
- Mice, Inbred C3H
- Mice, SCID
- Receptors, Fc/metabolism
- Rickettsia conorii/immunology
Collapse
Affiliation(s)
- Hui-Min Feng
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | |
Collapse
|
50
|
Drevets DA, Leenen PJM, Greenfield RA. Invasion of the central nervous system by intracellular bacteria. Clin Microbiol Rev 2004; 17:323-47. [PMID: 15084504 PMCID: PMC387409 DOI: 10.1128/cmr.17.2.323-347.2004] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Infection of the central nervous system (CNS) is a severe and frequently fatal event during the course of many diseases caused by microbes with predominantly intracellular life cycles. Examples of these include the facultative intracellular bacteria Listeria monocytogenes, Mycobacterium tuberculosis, and Brucella and Salmonella spp. and obligate intracellular microbes of the Rickettsiaceae family and Tropheryma whipplei. Unfortunately, the mechanisms used by intracellular bacterial pathogens to enter the CNS are less well known than those used by bacterial pathogens with an extracellular life cycle. The goal of this review is to elaborate on the means by which intracellular bacterial pathogens establish infection within the CNS. This review encompasses the clinical and pathological findings that pertain to the CNS infection in humans and includes experimental data from animal models that illuminate how these microbes enter the CNS. Recent experimental data showing that L. monocytogenes can invade the CNS by more than one mechanism make it a useful model for discussing the various routes for neuroinvasion used by intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Douglas A Drevets
- Department of Medicine, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma 73104, USA.
| | | | | |
Collapse
|