1
|
Cote CK, Mlynek KD, Klimko CP, Biryukov SS, Mou S, Hunter M, Rill NO, Dankmeyer JL, Miller JA, Talyansky Y, Davies ML, Meinig JM, Halasohoris SA, Gray AM, Spencer JL, Babyak AL, Hourihan MK, Curry BJ, Toothman RG, Ruiz SI, Zeng X, Ricks KM, Clements TL, Douglas CE, Ravulapalli S, Stefan CP, Shoemaker CJ, Elrod MG, Gee JE, Weiner ZP, Qiu J, Bozue JA, Twenhafel NA, DeShazer D. Virulence of Burkholderia pseudomallei ATS2021 Unintentionally Imported to United States in Aromatherapy Spray. Emerg Infect Dis 2024; 30:2056-2069. [PMID: 39320153 PMCID: PMC11431913 DOI: 10.3201/eid3010.240084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
In the United States in 2021, an outbreak of 4 cases of Burkholderia pseudomallei, the etiologic agent of melioidosis and a Tier One Select Agent (potential for deliberate misuse and subsequent harm), resulted in 2 deaths. The causative strain, B. pseudomallei ATS2021, was unintentionally imported into the United States in an aromatherapy spray manufactured in India. We established that ATS2021 represents a virulent strain of B. pseudomallei capable of robust formation of biofilm at physiologic temperatures that may contribute to virulence. By using mouse melioidosis models, we determined median lethal dose estimates and analyzed the bacteriologic and histopathologic characteristics of the organism, particularly the potential neurologic pathogenesis that is probably associated with the bimABm allele identified in B. pseudomallei strain ATS2021. Our data, combined with previous case reports and the identification of endemic B. pseudomallei strains in Mississippi, support the concept that melioidosis is emerging in the United States.
Collapse
|
2
|
Adler BL, Chung T, Rowe PC, Aucott J. Dysautonomia following Lyme disease: a key component of post-treatment Lyme disease syndrome? Front Neurol 2024; 15:1344862. [PMID: 38390594 PMCID: PMC10883079 DOI: 10.3389/fneur.2024.1344862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Dysautonomia, or dysfunction of the autonomic nervous system (ANS), may occur following an infectious insult and can result in a variety of debilitating, widespread, and often poorly recognized symptoms. Dysautonomia is now widely accepted as a complication of COVID-19 and is an important component of Post-Acute Sequelae of COVID-19 (PASC or long COVID). PASC shares many overlapping clinical features with other infection-associated chronic illnesses including Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) and Post-Treatment Lyme Disease Syndrome (PTLDS), suggesting that they may share common underlying mechanisms including autonomic dysfunction. Despite the recognition of this complication of Lyme disease in the care of patients with PTLD, there has been a scarcity of research in this field and dysautonomia has not yet been established as a complication of Lyme disease in the medical literature. In this review, we discuss the evidence implicating Borrelia burgdorferi as a cause of dysautonomia and the related symptoms, propose potential pathogenic mechanisms given our knowledge of Lyme disease and mechanisms of PASC and ME/CFS, and discuss the diagnostic evaluation and treatments of dysautonomia. We also outline gaps in the literature and priorities for future research.
Collapse
Affiliation(s)
- Brittany L Adler
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, United States
| | - Tae Chung
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C Rowe
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States
| | - John Aucott
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
3
|
Gadila SKG, Embers ME. Direct Detection of Borrelia Species in Tissues. Methods Mol Biol 2024; 2742:19-35. [PMID: 38165612 DOI: 10.1007/978-1-0716-3561-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Among the controversies in Lyme disease is the potential for Borrelia spirochetes to persist after guideline-directed antimicrobial therapy. Direct detection of the spirochetes has been essential to explore this phenomenon, given that the infection is often occult and infrequently observed in blood and other body fluids. In addition, the role of spirochetal infection has been examined in the etiology of neurodegenerative diseases through detection in affected tissues. In this chapter, we describe methodology to specifically identify Borrelia DNA, RNA, and intact organism (via protein) in tissue for studies of Lyme Borreliosis.
Collapse
Affiliation(s)
- Shiva Kumar Goud Gadila
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, New Orleans, LA, USA
| | - Monica E Embers
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, New Orleans, LA, USA.
| |
Collapse
|
4
|
Kulkarni A, Jozefiaková J, Bhide K, Mochnaćová E, Bhide M. Differential transcriptome response of blood brain barrier spheroids to neuroinvasive Neisseria and Borrelia. Front Cell Infect Microbiol 2023; 13:1326578. [PMID: 38179419 PMCID: PMC10766361 DOI: 10.3389/fcimb.2023.1326578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Background The blood-brain barrier (BBB), a highly regulated interface between the blood and the brain, prevents blood-borne substances and pathogens from entering the CNS. Nevertheless, pathogens like Neisseria meningitidis and Borrelia bavariensis can breach the BBB and infect the brain parenchyma. The self-assembling BBB-spheroids can simulate the cross talk occurring between the cells of the barrier and neuroinvasive pathogens. Methods BBB spheroids were generated by co-culturing human brain microvascular endothelial cells (hBMECs), pericytes and astrocytes. The BBB attributes of spheroids were confirmed by mapping the localization of cells, observing permeability of angiopep2 and non-permeability of dextran. Fluorescent Neisseria, Borrelia or E. coli (non-neuroinvasive) were incubated with spheroids to observe the adherence, invasion and spheroid integrity. Transcriptome analysis with NGS was employed to investigate the response of BBB cells to infections. Results hBMECs were localized throughout the spheroids, whereas pericytes and astrocytes were concentrated around the core. Within 1 hr of exposure, Neisseria and Borrelia adhered to spheroids, and their microcolonization increased from 5 to 24 hrs. Integrity of spheroids was compromised by both Neisseria and Borrelia, but not by E. coli infection. Transcriptome analysis revealed a significant change in the expression of 781 genes (467 up and 314 down regulated) in spheroids infected with Neisseria, while Borrelia altered the expression of 621 genes (225 up and 396 down regulated). The differentially expressed genes could be clustered into various biological pathways like cell adhesion, extracellular matrix related, metallothionines, members of TGF beta, WNT signaling, and immune response. Among the differentially expressed genes, 455 (48%) genes were inversely expressed during Neisseria and Borrelia infection. Conclusion The self-assembling spheroids were used to perceive the BBB response to neuroinvasive pathogens - Neisseria and Borrelia. Compromised integrity of spheroids during Neisseria and Borrelia infection as opposed to its intactness and non-adherence of E. coli (non-neuroinvasive) denotes the pathogen dependent fate of BBB. Genes categorized into various biological functions indicated weakened barrier properties of BBB and heightened innate immune response. Inverse expression of 48% genes commonly identified during Neisseria and Borrelia infection exemplifies unique response of BBB to varying neuropathogens.
Collapse
Affiliation(s)
- Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
- Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jana Jozefiaková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Evelína Mochnaćová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
- Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
5
|
Zhao C, Kuraji R, Ye C, Gao L, Radaic A, Kamarajan P, Taketani Y, Kapila YL. Nisin a probiotic bacteriocin mitigates brain microbiome dysbiosis and Alzheimer's disease-like neuroinflammation triggered by periodontal disease. J Neuroinflammation 2023; 20:228. [PMID: 37803465 PMCID: PMC10557354 DOI: 10.1186/s12974-023-02915-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023] Open
Abstract
INTRODUCTION Periodontitis-related oral microbial dysbiosis is thought to contribute to Alzheimer's disease (AD) neuroinflammation and brain amyloid production. Since probiotics can modulate periodontitis/oral dysbiosis, this study examined the effects of a probiotic/lantibiotic, nisin, in modulating brain pathology triggered by periodontitis. METHODS A polymicrobial mouse model of periodontal disease was used to evaluate the effects of this disease on brain microbiome dysbiosis, neuroinflammation, Alzheimer's-related changes, and nisin's therapeutic potential in this context. RESULTS 16S sequencing and real-time PCR data revealed that Nisin treatment mitigated the changes in the brain microbiome composition, diversity, and community structure, and reduced the levels of periodontal pathogen DNA in the brain induced by periodontal disease. Nisin treatment significantly decreased the mRNA expression of pro-inflammatory cytokines (Interleukin-1β/IL-1 β, Interleukin 6/IL-6, and Tumor Necrosis Factor α/TNF-α) in the brain that were elevated by periodontal infection. In addition, the concentrations of amyloid-β 42 (Aβ42), total Tau, and Tau (pS199) (445.69 ± 120.03, 1420.85 ± 331.40, 137.20 ± 36.01) were significantly higher in the infection group compared to the control group (193.01 ± 31.82, 384.27 ± 363.93, 6.09 ± 10.85), respectively. Nisin treatment markedly reduced the Aβ42 (261.80 ± 52.50), total Tau (865.37 ± 304.93), and phosphorylated Tau (82.53 ± 15.77) deposition in the brain of the infection group. DISCUSSION Nisin abrogation of brain microbiome dysbiosis induces beneficial effects on AD-like pathogenic changes and neuroinflammation, and thereby may serve as a potential therapeutic for periodontal-dysbiosis-related AD.
Collapse
Affiliation(s)
- Chuanjiang Zhao
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
| | - Ryutaro Kuraji
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, 102-8159, Japan
| | - Changchang Ye
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, West China School of Stomatology, National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610093, China
| | - Li Gao
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
| | - Allan Radaic
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Yoshimasa Taketani
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, 350-0283, Japan
| | - Yvonne L Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA.
- Section of Biosystems and Function, Section of Periodontology, UCLA School of Dentistry, 10833 Le Conte Ave, Box 951668, Los Angeles, CA, 90095-1668, USA.
| |
Collapse
|
6
|
Adkison H, Embers ME. Lyme disease and the pursuit of a clinical cure. Front Med (Lausanne) 2023; 10:1183344. [PMID: 37293310 PMCID: PMC10244525 DOI: 10.3389/fmed.2023.1183344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023] Open
Abstract
Lyme disease, caused by the spirochete Borrelia burgdorferi, is the most common vector-borne illness in the United States. Many aspects of the disease are still topics of controversy within the scientific and medical communities. One particular point of debate is the etiology behind antibiotic treatment failure of a significant portion (10-30%) of Lyme disease patients. The condition in which patients with Lyme disease continue to experience a variety of symptoms months to years after the recommended antibiotic treatment is most recently referred to in the literature as post treatment Lyme disease syndrome (PTLDS) or just simply post treatment Lyme disease (PTLD). The most commonly proposed mechanisms behind treatment failure include host autoimmune responses, long-term sequelae from the initial Borrelia infection, and persistence of the spirochete. The aims of this review will focus on the in vitro, in vivo, and clinical evidence that either validates or challenges these mechanisms, particularly with regard to the role of the immune response in disease and resolution of the infection. Next generation treatments and research into identifying biomarkers to predict treatment responses and outcomes for Lyme disease patients are also discussed. It is essential that definitions and guidelines for Lyme disease evolve with the research to translate diagnostic and therapeutic advances to patient care.
Collapse
Affiliation(s)
| | - Monica E. Embers
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA, United States
| |
Collapse
|
7
|
Implications of Microorganisms in Alzheimer's Disease. Curr Issues Mol Biol 2022; 44:4584-4615. [PMID: 36286029 PMCID: PMC9600878 DOI: 10.3390/cimb44100314] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a deadly brain degenerative disorder that leads to brain shrinkage and dementia. AD is manifested with hyperphosphorylated tau protein levels and amyloid beta (Aβ) peptide buildup in the hippocampus and cortex regions of the brain. The nervous tissue of AD patients also contains fungal proteins and DNA which are linked to bacterial infections, suggesting that polymicrobial infections also occur in the brains of those with AD. Both immunohistochemistry and next-generation sequencing (NGS) techniques were employed to assess fungal and bacterial infections in the brain tissue of AD patients and non-AD controls, with the most prevalent fungus genera detected in AD patients being Alternaria, Botrytis, Candida, and Malassezia. Interestingly, Fusarium was the most common genus detected in the control group. Both AD patients and controls were also detectable for Proteobacteria, followed by Firmicutes, Actinobacteria, and Bacteroides for bacterial infection. At the family level, Burkholderiaceae and Staphylococcaceae exhibited higher levels in the brains of those with AD than the brains of the control group. Accordingly, there is thought to be a viscous cycle of uncontrolled neuroinflammation and neurodegeneration in the brain, caused by agents such as the herpes simplex virus type 1 (HSV1), Chlamydophilapneumonia, and Spirochetes, and the presence of apolipoprotein E4 (APOE4), which is associated with an increased proinflammatory response in the immune system. Systemic proinflammatory cytokines are produced by microorganisms such as Cytomegalovirus, Helicobacter pylori, and those related to periodontal infections. These can then cross the blood–brain barrier (BBB) and lead to the onset of dementia. Here, we reviewed the relationship between the etiology of AD and microorganisms (such as bacterial pathogens, Herpesviridae viruses, and periodontal pathogens) according to the evidence available to understand the pathogenesis of AD. These findings might guide a targeted anti-inflammatory therapeutic approach to AD.
Collapse
|
8
|
Courtier A, Potheret D, Giannoni P. Environmental bacteria as triggers to brain disease: Possible mechanisms of toxicity and associated human risk. Life Sci 2022; 304:120689. [DOI: 10.1016/j.lfs.2022.120689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
|
9
|
Landry RL, Embers ME. Does Dementia Have a Microbial Cause? NEUROSCI 2022; 3:262-283. [PMID: 39483362 PMCID: PMC11523730 DOI: 10.3390/neurosci3020019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/03/2024] Open
Abstract
The potential contribution of pathogenic microbes to dementia-inducing disease is a subject of considerable importance. Alzheimer's disease (AD) is a neurocognitive disease that slowly destroys brain function, leading to cognitive decline and behavioral and psychiatric disorders. The histopathology of AD is associated with neuronal loss and progressive synaptic dysfunction, accompanied by the deposition of amyloid-β (Aβ) peptide in the form of parenchymal plaques and abnormal aggregated tau protein in the form of neurofibrillary tangles. Observational, epidemiological, experimental, and pathological studies have generated evidence for the complexity and possible polymicrobial causality in dementia-inducing diseases. The AD pathogen hypothesis states that pathogens and microbes act as triggers, interacting with genetic factors to initiate the accumulation of Aβ, hyperphosphorylated tau protein (p-tau), and inflammation in the brain. Evidence indicates that Borrelia sp., HSV-1, VZV (HHV-2), HHV-6/7, oral pathogens, Chlamydophila pneumoniae, and Candida albicans can infect the central nervous system (CNS), evade the immune system, and consequently prevail in the AD brain. Researchers have made significant progress in understanding the multifactorial and overlapping factors that are thought to take part in the etiopathogenesis of dementia; however, the cause of AD remains unclear.
Collapse
Affiliation(s)
- Remi L Landry
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| | - Monica E Embers
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| |
Collapse
|
10
|
Parthasarathy G, Gadila SKG. Neuropathogenicity of non-viable Borrelia burgdorferi ex vivo. Sci Rep 2022; 12:688. [PMID: 35027599 PMCID: PMC8758786 DOI: 10.1038/s41598-021-03837-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Even after appropriate treatment, a proportion of Lyme disease patients suffer from a constellation of symptoms, collectively called Post-Treatment Lyme Disease Syndrome (PTLDS). Brain PET scan of patients with PTLDS have demonstrated likely glial activation indicating persistent neuroinflammatory processes. It is possible that unresolved bacterial remnants can continue to cause neuroinflammation. In previous studies, we have shown that non-viable Borrelia burgdorferi can induce neuroinflammation and apoptosis in an oligodendrocyte cell line. In this follow-up study, we analyze the effect of sonicated remnants of B. burgdorferi on primary rhesus frontal cortex (FC) and dorsal root ganglion (DRG) explants. Five FC and three DRG tissue fragments from rhesus macaques were exposed to sonicated B. burgdorferi and analyzed for 26 inflammatory mediators. Live bacteria and medium alone served as positive and negative control, respectively. Tissues were also analyzed for cell types mediating inflammation and overall apoptotic changes. Non-viable B. burgdorferi induced significant levels of several inflammatory mediators in both FC and DRG, similar to live bacteria. However, the levels induced by non-viable B. burgdorferi was often (several fold) higher than those induced by live ones, especially for IL-6, CXCL8 and CCL2. This effect was also more profound in the FC than in the DRG. Although the levels often differed, both live and dead fragments induced the same mediators, with significant overlap between FC and DRG. In the FC, immunohistochemical staining for several inflammatory mediators showed the presence of multiple mediators in astrocytes, followed by microglia and oligodendrocytes, in response to bacterial remnants. Staining was also seen in endothelial cells. In the DRG, chemokine/cytokine staining was predominantly seen in S100 positive (glial) cells. B. burgdorferi remnants also induced significant levels of apoptosis in both the FC and DRG. Apoptosis was confined to S100 + cells in the DRG while distinct neuronal apoptosis was also detected in most FC tissues in response to sonicated bacteria. Non-viable B. burgdorferi can continue to be neuropathogenic to both CNS and PNS tissues with effects likely more profound in the former. Persistence of remnant-induced neuroinflammatory processes can lead to long term health consequences.
Collapse
Affiliation(s)
- Geetha Parthasarathy
- Division of Immunology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA. .,Tulane National Primate Research Center, 18703, Three rivers Road, Room 109, Covington, LA, 70433, USA.
| | - Shiva Kumar Goud Gadila
- Division of Immunology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA
| |
Collapse
|
11
|
Hammel JH, Cook SR, Belanger MC, Munson JM, Pompano RR. Modeling Immunity In Vitro: Slices, Chips, and Engineered Tissues. Annu Rev Biomed Eng 2021; 23:461-491. [PMID: 33872520 PMCID: PMC8277680 DOI: 10.1146/annurev-bioeng-082420-124920] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Modeling immunity in vitro has the potential to be a powerful tool for investigating fundamental biological questions, informing therapeutics and vaccines, and providing new insight into disease progression. There are two major elements to immunity that are necessary to model: primary immune tissues and peripheral tissues with immune components. Here, we systematically review progress made along three strategies to modeling immunity: ex vivo cultures, which preserve native tissue structure; microfluidic devices, which constitute a versatile approach to providing physiologically relevant fluid flow and environmental control; and engineered tissues, which provide precise control of the 3D microenvironment and biophysical cues. While many models focus on disease modeling, more primary immune tissue models are necessary to advance the field. Moving forward, we anticipate that the expansion of patient-specific models may inform why immunity varies from patient to patient and allow for the rapid comprehension and treatment of emerging diseases, such as coronavirus disease 2019.
Collapse
Affiliation(s)
- Jennifer H Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Sophie R Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Maura C Belanger
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Jennifer M Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA;
- Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| |
Collapse
|
12
|
Ford L, Tufts DM. Lyme Neuroborreliosis: Mechanisms of B. burgdorferi Infection of the Nervous System. Brain Sci 2021; 11:brainsci11060789. [PMID: 34203671 PMCID: PMC8232152 DOI: 10.3390/brainsci11060789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Lyme borreliosis is the most prevalent tick-borne disease in the United States, infecting ~476,000 people annually. Borrelia spp. spirochetal bacteria are the causative agents of Lyme disease in humans and are transmitted by Ixodes spp ticks. Clinical manifestations vary depending on which Borrelia genospecies infects the patient and may be a consequence of distinct organotropism between species. In the US, B. burgdorferi sensu stricto is the most commonly reported genospecies and infection can manifest as mild to severe symptoms. Different genotypes of B. burgdorferi sensu stricto may be responsible for causing varying degrees of clinical manifestations. While the majority of Lyme borreliae-infected patients fully recover with antibiotic treatment, approximately 15% of infected individuals experience long-term neurological and psychological symptoms that are unresponsive to antibiotics. Currently, long-term antibiotic treatment remains the only FDA-approved option for those suffering from these chronic effects. Here, we discuss the current knowledge pertaining to B. burgdorferi sensu stricto infection in the central nervous system (CNS), termed Lyme neuroborreliosis (LNB), within North America and specifically the United States. We explore the molecular mechanisms of spirochete entry into the brain and the role B. burgdorferi sensu stricto genotypes play in CNS infectivity. Understanding infectivity can provide therapeutic targets for LNB treatment and offer public health understanding of the B. burgdorferi sensu stricto genotypes that cause long-lasting symptoms.
Collapse
Affiliation(s)
- Lenzie Ford
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
- Correspondence: (L.F.); (D.M.T.)
| | - Danielle M. Tufts
- Infectious Diseases and Microbiology Department, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (L.F.); (D.M.T.)
| |
Collapse
|
13
|
Li L, Luo L, Chen T, Cao W, Xu X, Zhang Y, Yue P, Fan Y, Chen J, Liu M, Ma M, Tao L, Peng Y, Dong Y, Li B, Luo S, Kong J, Zhou G, Wen S, Liu A, Bao F. Proteomic Analysis of Rhesus Macaque Brain Explants Treated With Borrelia burgdorferi Identifies Host GAP-43 as a Potential Factor Associated With Lyme Neuroborreliosis. Front Cell Infect Microbiol 2021; 11:647662. [PMID: 34178719 PMCID: PMC8224226 DOI: 10.3389/fcimb.2021.647662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/25/2021] [Indexed: 12/02/2022] Open
Abstract
Background Lyme neuroborreliosis (LNB) is one of the most dangerous manifestations of Lyme disease, but the pathogenesis and inflammatory mechanisms are not fully understood. Methods Cultured explants from the frontal cortex of rhesus monkey brain (n=3) were treated with live Borrelia burgdorferi (Bb) or phosphate-buffered saline (PBS) for 6, 12, and 24 h. Total protein was collected for sequencing and bioinformatics analysis. In addition, changes in protein expression in the explants over time following Bb treatment were screened. Results We identified 1237 differentially expressed proteins (DEPs; fold change ≥1.5 or ≤0.67, P-value ≤0.05). One of these, growth-associated protein 43 (GAP-43), was highly expressed at all time points in the explants. The results of the protein-protein interaction network analysis of DEPs suggested that GAP-43 plays a role in the neuroinflammation associated with LNB. In HMC3 cells incubated with live Bb or PBS for 6, 12, and 24 h, real-time PCR and western blot analyses confirmed the increase of GAP-43 mRNA and protein, respectively. Conclusions Elevated GAP-43 expression is a potential marker for LNB that may be useful for diagnosis or treatment.
Collapse
Affiliation(s)
- Lianbao Li
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lisha Luo
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Taigui Chen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Wenjing Cao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Xin Xu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Yu Zhang
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Peng Yue
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Yuxin Fan
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Jingjing Chen
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Meixiao Liu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Mingbiao Ma
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lvyan Tao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Yun Peng
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Yan Dong
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Bingxue Li
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Suyi Luo
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Jing Kong
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Guozhong Zhou
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Shiyuan Wen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Aihua Liu
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China.,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming/Kunming Medical University, Kunming, China.,The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Fukai Bao
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming/Kunming Medical University, Kunming, China.,The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| |
Collapse
|
14
|
Gadila SKG, Rosoklija G, Dwork AJ, Fallon BA, Embers ME. Detecting Borrelia Spirochetes: A Case Study With Validation Among Autopsy Specimens. Front Neurol 2021; 12:628045. [PMID: 34040573 PMCID: PMC8141553 DOI: 10.3389/fneur.2021.628045] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 01/30/2023] Open
Abstract
The complex etiology of neurodegenerative disease has prompted studies on multiple mechanisms including genetic predisposition, brain biochemistry, immunological responses, and microbial insult. In particular, Lyme disease is often associated with neurocognitive impairment with variable manifestations between patients. We sought to develop methods to reliably detect Borrelia burgdorferi, the spirochete bacteria responsible for Lyme disease, in autopsy specimens of patients with a history of neurocognitive disease. In this report, we describe the use of multiple molecular detection techniques for this pathogen and its application to a case study of a Lyme disease patient. The patient had a history of Lyme disease, was treated with antibiotics, and years later developed chronic symptoms including dementia. The patient's pathology and clinical case description was consistent with Lewy body dementia. B. burgdorferi was identified by PCR in several CNS tissues and by immunofluorescent staining in the spinal cord. These studies offer proof of the principle that persistent infection with the Lyme disease spirochete may have lingering consequences on the CNS.
Collapse
Affiliation(s)
- Shiva Kumar Goud Gadila
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA, United States
| | - Gorazd Rosoklija
- Department of Psychiatry, Columbia University, New York, NY, United States.,Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, United States
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University, New York, NY, United States.,Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, United States.,Macedonian Academy of Sciences and Arts, Skopje, Macedonia.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Brian A Fallon
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Monica E Embers
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA, United States
| |
Collapse
|
15
|
Thompson D, Watt JA, Brissette CA. Host transcriptome response to Borrelia burgdorferi sensu lato. Ticks Tick Borne Dis 2020; 12:101638. [PMID: 33360384 DOI: 10.1016/j.ttbdis.2020.101638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
The host immune response to infection is a well-coordinated system of innate and adaptive immune cells working in concert to prevent the colonization and dissemination of a pathogen. While this typically leads to a beneficial outcome and the suppression of disease pathogenesis, the Lyme borreliosis bacterium, Borrelia burgdorferi sensu lato, can elicit an immune profile that leads to a deleterious state. As B. burgdorferi s.l. produces no known toxins, it is suggested that the immune and inflammatory response of the host are responsible for the manifestation of symptoms, including flu-like symptoms, musculoskeletal pain, and cognitive disorders. The past several years has seen a substantial increase in the use of microarray and sequencing technologies to investigate the transcriptome response induced by B. burgdorferi s.l., thus enabling researchers to identify key factors and pathways underlying the pathophysiology of Lyme borreliosis. In this review we present the major host transcriptional outcomes induced by the bacterium across several studies and discuss the overarching theme of the host inflammatory and immune response, and how it influences the pathology of Lyme borreliosis.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| | - John A Watt
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| | - Catherine A Brissette
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| |
Collapse
|
16
|
Abstract
Lyme disease (Lyme borreliosis) is a tick-borne, zoonosis of adults and children caused by genospecies of the Borrelia burgdorferi sensu lato complex. The ailment, widespread throughout the Northern Hemisphere, continues to increase globally due to multiple environmental factors, coupled with increased incursion of humans into habitats that harbor the spirochete. B. burgdorferi sensu lato is transmitted by ticks from the Ixodes ricinus complex. In North America, B. burgdorferi causes nearly all infections; in Europe, B. afzelii and B. garinii are most associated with human disease. The spirochete's unusual fragmented genome encodes a plethora of differentially expressed outer surface lipoproteins that play a seminal role in the bacterium's ability to sustain itself within its enzootic cycle and cause disease when transmitted to its incidental human host. Tissue damage and symptomatology (i.e., clinical manifestations) result from the inflammatory response elicited by the bacterium and its constituents. The deposition of spirochetes into human dermal tissue generates a local inflammatory response that manifests as erythema migrans (EM), the hallmark skin lesion. If treated appropriately and early, the prognosis is excellent. However, in untreated patients, the disease may present with a wide range of clinical manifestations, most commonly involving the central nervous system, joints, or heart. A small percentage (~10%) of patients may go on to develop a poorly defined fibromyalgia-like illness, post-treatment Lyme disease (PTLD) unresponsive to prolonged antimicrobial therapy. Below we integrate current knowledge regarding the ecologic, epidemiologic, microbiologic, and immunologic facets of Lyme disease into a conceptual framework that sheds light on the disorder that healthcare providers encounter.
Collapse
Affiliation(s)
- Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT 06030, USA
- Department of Pediatrics, UConn Health, Farmington, CT 06030, USA
- Departments of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
- Departments of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Klemen Strle
- Division of Infectious Diseases, Wadsworth Center, NY Department of Health, Albany NY, 12208, USA
| | - Jacob E. Lemieux
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
17
|
Ding Z, Sun L, Bi Y, Zhang Y, Yue P, Xu X, Cao W, Luo L, Chen T, Li L, Ji Z, Jian M, Lu L, Abi ME, Liu A, Bao F. Integrative Transcriptome and Proteome Analyses Provide New Insights Into the Interaction Between Live Borrelia burgdorferi and Frontal Cortex Explants of the Rhesus Brain. J Neuropathol Exp Neurol 2020; 79:518-529. [PMID: 32196082 DOI: 10.1093/jnen/nlaa015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/03/2020] [Accepted: 02/13/2020] [Indexed: 01/01/2023] Open
Abstract
Borrelia burgdorferi (Bb), which is neurotropic, can attack the central nervous system (CNS), leading to the development of various neurologic symptoms. The pathogenesis of Lyme neuroborreliosis (LNB) remains poorly understood. Presently, there is a lack of knowledge of the changes in mRNA and proteins in the CNS following early disseminated Lyme disease. Explants from the frontal cortex of 3 rhesus brains were incubated with medium alone or with medium containing live Bb for 6, 12, or 24 hours. Then, we analyzed identified mRNA and proteins in the frontal cortex tissues, allowing for an in-depth view of the transcriptome and proteome for a macroscopic and unbiased understanding of early disseminated Lyme disease in the brain. Through bioinformatics analysis, a complex network of enriched pathways that were mobilized during the progression of Lyme spirochete infection was described. Furthermore, based on the analysis of omics data, translational regulation, glycosaminoglycan/proteoglycan-binding activity in colonization and dissemination to tissues, disease-associated genes, and synaptic function were enriched, which potentially play a role in pathogenesis during the interaction between frontal cortex tissues and spirochetes. These integrated omics results provide unbiased and comprehensive information for the further understanding of the molecular mechanisms of LNB.
Collapse
Affiliation(s)
- Zhe Ding
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Luyun Sun
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities
| | - Yunfeng Bi
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities
| | - Yu Zhang
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Peng Yue
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Xin Xu
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Biochemistry and Molecular Biology, Kunming Medical University
| | - Wenjing Cao
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Biochemistry and Molecular Biology, Kunming Medical University
| | - Lisha Luo
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Biochemistry and Molecular Biology, Kunming Medical University
| | - Taigui Chen
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Lianbao Li
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Zhenhua Ji
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Miaomiao Jian
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Biochemistry and Molecular Biology, Kunming Medical University
| | - Lihong Lu
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities
| | - Manzama-Esso Abi
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Department of Microbiology and Immunology
| | - Aihua Liu
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Yunnan Province Key Laboratory for Children's Major Diseases Research, The Children's Hospital of Kunming.,Department of Biochemistry and Molecular Biology, Kunming Medical University.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Fukai Bao
- From the Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities.,Yunnan Province Key Laboratory for Children's Major Diseases Research, The Children's Hospital of Kunming.,Department of Microbiology and Immunology.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| |
Collapse
|
18
|
Zubcevik N, Mao C, Wang QM, Bose EL, Octavien RN, Crandell D, Wood LJ. Symptom Clusters and Functional Impairment in Individuals Treated for Lyme Borreliosis. Front Med (Lausanne) 2020; 7:464. [PMID: 32974369 PMCID: PMC7472530 DOI: 10.3389/fmed.2020.00464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/13/2020] [Indexed: 11/17/2022] Open
Abstract
Context: Persistent fatigue, pain, and neurocognitive impairment are common in individuals following treatment for Lyme borreliosis (LB). Poor sleep, depression, visual disturbance, and sensory neuropathies have also been reported. The cause of these symptoms is unclear, and widely accepted effective treatment strategies are lacking. Objectives: To identify symptom clusters in people with persistent symptoms previously treated for LB and to examine the relationship between symptom severity and perceived disability. Methods: This was a retrospective chart review of individuals with a history of treatment of LB referred to The Dean Center for Tick-Borne Illness at Spaulding Rehabilitation Hospital between 2015 and 2018 (n = 270) because of persistent symptoms. Symptoms and functional impairment were collected using the General Symptom Questionnaire-30 (GSQ-30), and the Sheehan Disability Scale. Clinical tests were conducted to evaluate for tick-borne co-infections and to rule out medical disorders that could mimic LB symptomatology. Exploratory factor analysis was performed to identify symptom clusters. Results: Five symptom clusters were identified. Each cluster was assigned a name to reflect the possible underlying etiology and was based on the majority of the symptoms in the cluster: the neuropathy symptom cluster, sleep-fatigue symptom cluster, migraine symptom cluster, cognitive symptom cluster, and mood symptom cluster. Symptom severity for each symptom cluster was positively associated with global functional impairment (p < 0.001). Conclusion: Identifying the interrelationship between symptoms in post-treatment LB in a cluster can aid in the identification of the etiological basis of these symptoms and could lead to more effective symptom management strategies. Key Message: This article describes symptom clusters in individuals with a history of Lyme borreliosis. Five clusters were identified: sleep-fatigue, neuropathy, migraine-like, cognition, and mood clusters. Identifying the interrelationship between symptoms in each of the identified clusters could aid in more effective symptom management through identifying triggering symptoms or an underlying etiology.
Collapse
Affiliation(s)
- Nevena Zubcevik
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, United States.,The Dean Center for Tickborne Illness, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States.,Invisible International, Cambridge, MA, United States
| | - Charlotte Mao
- The Dean Center for Tickborne Illness, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States.,Massachusetts General Hospital, Department of Pediatric Infectious Diseases, Boston, MA, United States
| | - Qing Mei Wang
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, United States.,Stroke Biological Recovery Laboratory, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Eliezer L Bose
- Stroke Biological Recovery Laboratory, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Rose Nadlyne Octavien
- The Dean Center for Tickborne Illness, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - David Crandell
- The Dean Center for Tickborne Illness, Spaulding Research Institute, Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Lisa J Wood
- Massachusetts General Hospital, Institute for Health Professions, School of Nursing, Charlestown, MA, United States.,William F. Connell School of Nursing, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
19
|
Scott JD. Presentation of Acrodermatitis Chronica Atrophicans Rashes on Lyme Disease Patients in Canada. Healthcare (Basel) 2020; 8:E157. [PMID: 32512846 PMCID: PMC7349802 DOI: 10.3390/healthcare8020157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 01/11/2023] Open
Abstract
Lyme disease (Lyme borreliosis) is a complex multisystem illness with varying clinical manifestations. This tick-borne zoonosis is caused by the spirochetal bacterium, Borrelia burgdorferi sensu lato (Bbsl) and, worldwide, presents with at least 20 different types of rashes. Certain cutaneous rashes are inherently interconnected to various stages of Lyme disease. In this study, five Canadian Lyme disease patients from a multi-age range presented various phases of the acrodermatitis chronica atrophicans (ACA) rash. In each case of ACA, the underlying etiological pathogen was the Lyme disease spirochete. Although ACA rashes are normally found on the lower extremities, this study illustrates that ACA rashes are not directly correlated with a tick bite, geographic area, age, Bbsl genospecies, exercise, or any given surface area of the body. Case 4 provides confirmation for an ACA rash and gestational Lyme disease. One patient (Case 5) puts forth a Bbsl and Bartonella sp. co-infection with a complex ACA rash. This study documents ACA rashes on Lyme disease patients for the first time in Canada.
Collapse
Affiliation(s)
- John D Scott
- International Lyme and Associated Diseases Society, 2 Wisconsin Circle, Suite 700, Chevy Chase, MD 20815-7007, USA
| |
Collapse
|
20
|
Effect of Borrelia burgdorferi Outer Membrane Vesicles on Host Oxidative Stress Response. Antibiotics (Basel) 2020; 9:antibiotics9050275. [PMID: 32466166 PMCID: PMC7277464 DOI: 10.3390/antibiotics9050275] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Outer membrane vesicles (OMVs) are spherical bodies containing proteins and nucleic acids that are released by Gram-negative bacteria, including Borrelia burgdorferi, the causative agent of Lyme disease. The functional relationship between B. burgdorferi OMVs and host neuron homeostasis is not well understood. The objective of this study was to examine how B. burgdorferi OMVs impact the host cell environment. First, an in vitro model was established by co-culturing human BE2C neuroblastoma cells with B. burgdorferi B31. B. burgdorferi was able to invade BE2C cells within 24 h. Despite internalization, BE2C cell viability and levels of apoptosis remained unchanged, but resulted in dramatically increased production of MCP-1 and MCP-2 cytokines. Elevated secretion of MCP-1 has previously been associated with changes in oxidative stress. BE2C cell mitochondrial superoxides were reduced as early as 30 min after exposure to B. burgdorferi and OMVs. To rule out whether BE2C cell antioxidant response is the cause of decline in superoxides, superoxide dismutase 2 (SOD2) gene expression was assessed. SOD2 expression was reduced upon exposure to B. burgdorferi, suggesting that B. burgdorferi might be responsible for superoxide reduction. These results suggest that B. burgdorferi modulates cell antioxidant defense and immune system reaction in response to the bacterial infection. In summary, these results show that B. burgdorferi OMVs serve to directly counter superoxide production in BE2C neurons, thereby 'priming' the host environment to support B. burgdorferi colonization.
Collapse
|
21
|
Monitoring of Nesting Songbirds Detects Established Population of Blacklegged Ticks and Associated Lyme Disease Endemic Area in Canada. Healthcare (Basel) 2020; 8:healthcare8010059. [PMID: 32183171 PMCID: PMC7151351 DOI: 10.3390/healthcare8010059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
This study provides a novel method of documenting established populations of bird-feeding ticks. Single populations of the blacklegged tick, Ixodes scapularis, and the rabbit tick, Haemaphysalis leporispalustris, were revealed in southwestern Québec, Canada. Blacklegged tick nymphs and, similarly, larval and nymphal rabbit ticks were tested for the Lyme disease bacterium, Borrelia burgdorferi sensu lato (Bbsl), using PCR and the flagellin (flaB) gene, and 14 (42%) of 33 of blacklegged tick nymphs tested were positive. In contrast, larval and nymphal H. leporsipalustris ticks were negative for Bbsl. The occurrence of Bbsl in I. scapularis nymphs brings to light the presence of a Lyme disease endemic area at this songbird nesting locality. Because our findings denote that this area is a Lyme disease endemic area, and I. scapularis is a human-biting tick, local residents and outdoor workers must take preventive measures to avoid tick bites. Furthermore, local healthcare practitioners must include Lyme disease in their differential diagnosis.
Collapse
|
22
|
Scott JD, Clark KL, Coble NM, Ballantyne TR. Detection and Transstadial Passage of Babesia Species and Borrelia burgdorferi Sensu Lato in Ticks Collected from Avian and Mammalian Hosts in Canada. Healthcare (Basel) 2019; 7:E155. [PMID: 31810270 PMCID: PMC6955799 DOI: 10.3390/healthcare7040155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 11/17/2022] Open
Abstract
Lyme disease and human babesiosis are the most common tick-borne zoonoses in the Temperate Zone of North America. The number of infected patients has continued to rise globally, and these zoonoses pose a major healthcare threat. This tick-host-pathogen study was conducted to test for infectious microbes associated with Lyme disease and human babesiosis in Canada. Using the flagellin (flaB) gene, three members of the Borrelia burgdorferi sensu lato (Bbsl) complex were detected, namely a Borrelia lanei-like spirochete, Borrelia burgdorferi sensu stricto (Bbss), and a distinct strain that may represent a separate Bbsl genospecies. This novel Bbsl strain was detected in a mouse tick, Ixodes muris, collected from a House Wren, Troglodytes aedon, in Quebec during the southward fall migration. The presence of Bbsl in bird-feeding larvae of I. muris suggests reservoir competency in three passerines (i.e., Common Yellowthroat, House Wren, Magnolia Warbler). Based on the 18S ribosomal RNA (rRNA) gene, three Babesia species (i.e., Babesia divergens-like, Babesia microti, Babesia odocoilei) were detected in field-collected ticks. Not only was B. odocoilei found in songbird-derived ticks, this piroplasm was apparent in adult questing blacklegged ticks, Ixodes scapularis, in southern Canada. By allowing live, engorged ticks to molt, we confirm the transstadial passage of Bbsl in I. muris and B. odocoilei in I. scapularis. Bbss and Babesia microti were detected concurrently in a groundhog tick, Ixodes cookei, in Western Ontario. In Alberta, a winter tick, Dermacentor albipictus, which was collected from a moose, Alces alces, tested positive for Bbss. Notably, a B. divergens-like piroplasm was detected in a rabbit tick, Haemaphysalis leporispalustris, collected from an eastern cottontail in southern Manitoba; this Babesia species is a first-time discovery in Canada. This rabbit tick was also co-infected with Borrelia lanei-like spirochetes, which constitutes a first in Canada. Overall, five ticks were concurrently infected with Babesia and Bbsl pathogens and, after the molt, could potentially co-infect humans. Notably, we provide the first authentic report of I. scapularis ticks co-infected with Bbsl and B. odocoilei in Canada. The full extent of infectious microorganisms transmitted to humans by ticks is not fully elucidated, and clinicians need to be aware of the complexity of these tick-transmitted enzootic agents on human health. Diagnosis and treatment must be administered by those with accredited medical training in tick-borne zoonosis.
Collapse
Affiliation(s)
- John D. Scott
- International Lyme and Associated Diseases Society, 2 Wisconsin Circle, Suite 700, Chevy Chase, MD 20815-7007, USA
| | - Kerry L. Clark
- Environmental Epidemiology Research Laboratory, Department of Public Health, University of North Florida, Jacksonville, FL 32224, USA; (K.L.C.); (N.M.C.); (T.R.B.)
| | - Nikki M. Coble
- Environmental Epidemiology Research Laboratory, Department of Public Health, University of North Florida, Jacksonville, FL 32224, USA; (K.L.C.); (N.M.C.); (T.R.B.)
| | - Taylor R. Ballantyne
- Environmental Epidemiology Research Laboratory, Department of Public Health, University of North Florida, Jacksonville, FL 32224, USA; (K.L.C.); (N.M.C.); (T.R.B.)
| |
Collapse
|
23
|
Aguayo S, Schuh CMAP, Vicente B, Aguayo LG. Association between Alzheimer's Disease and Oral and Gut Microbiota: Are Pore Forming Proteins the Missing Link? J Alzheimers Dis 2019; 65:29-46. [PMID: 30040725 DOI: 10.3233/jad-180319] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition affecting millions of people worldwide. It is associated with cerebral amyloid-β (Aβ) plaque deposition in the brain, synaptic disconnection, and subsequent progressive neuronal death. Although considerable progress has been made to elucidate the pathogenesis of AD, the specific causes of the disease remain highly unknown. Recent research has suggested a potential association between certain infectious diseases and dementia, either directly due to bacterial brain invasion and toxin production, or indirectly by modulating the immune response. Therefore, in the present review we focus on the emerging issues of bacterial infection and AD, including the existence of antimicrobial peptides having pore-forming properties that act in a similar way to pores formed by Aβ in a variety of cell membranes. Special focus is placed on oral bacteria and biofilms, and on the potential mechanisms associating bacterial infection and toxin production in AD. The role of bacterial outer membrane vesicles on the transport and delivery of toxins as well as porins to the brain is also discussed. Aβ has shown to possess antimicrobial activity against several bacteria, and therefore could be upregulated as a response to bacteria and bacterial toxins in the brain. Although further research is needed, we believe that the control of biofilm-mediated diseases could be an important potential prevention mechanism for AD development.
Collapse
|
24
|
Djokic V, Akoolo L, Primus S, Schlachter S, Kelly K, Bhanot P, Parveen N. Protozoan Parasite Babesia microti Subverts Adaptive Immunity and Enhances Lyme Disease Severity. Front Microbiol 2019; 10:1596. [PMID: 31354683 PMCID: PMC6635642 DOI: 10.3389/fmicb.2019.01596] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Lyme disease is the most prominent tick-borne disease in the United States. Co-infections with the tick-transmitted pathogens Babesia microti and Borrelia burgdorferi sensu stricto are becoming a serious health problem. B. burgdorferi is an extracellular spirochete that causes Lyme disease while B. microti is a protozoan that infects erythrocytes and causes babesiosis. Testing of donated blood for Babesia species is not currently mandatory due to unavailability of an FDA approved test. Transmission of this protozoan by blood transfusion often results in high morbidity and mortality in recipients. Infection of C3H/HeJ mice with B. burgdorferi and B. microti individually results in inflammatory Lyme disease and display of human babesiosis-like symptoms, respectively. Here we use this mouse model to provide a detailed investigation of the reciprocal influence of the two pathogens on each other during co-infection. We show that B. burgdorferi infection attenuates parasitemia in mice while B. microti subverts the splenic immune response, such that a marked decrease in splenic B and T cells, reduction in antibody levels and diminished functional humoral immunity, as determined by spirochete opsonophagocytosis, are observed in co-infected mice compared to only B. burgdorferi infected mice. Furthermore, immunosuppression by B. microti in co-infected mice showed an association with enhanced Lyme disease manifestations. This study demonstrates the effect of only simultaneous infection by B. burgdorferi and B. microti on each pathogen, immune response and on disease manifestations with respect to infection by the spirochete and the parasite. In our future studies, we will examine the overall effects of sequential infection by these pathogens on host immune responses and disease outcomes.
Collapse
Affiliation(s)
- Vitomir Djokic
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Lavoisier Akoolo
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Shekerah Primus
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Samantha Schlachter
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Kathleen Kelly
- Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| | - Purnima Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
25
|
Ding Z, Ma M, Tao L, Peng Y, Han Y, Sun L, Dai X, Ji Z, Bai R, Jian M, Chen T, Luo L, Wang F, Bi Y, Liu A, Bao F. Rhesus Brain Transcriptomic Landscape in an ex vivo Model of the Interaction of Live Borrelia Burgdorferi With Frontal Cortex Tissue Explants. Front Neurosci 2019; 13:651. [PMID: 31316336 PMCID: PMC6610209 DOI: 10.3389/fnins.2019.00651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022] Open
Abstract
Lyme neuroborreliosis (LNB) is the most dangerous manifestation of Lyme disease caused by the spirochete Borrelia burgdorferi which can reach the central nervous system most commonly presenting with lymphocytic meningitis; however, the molecular basis for neuroborreliosis is still poorly understood. We incubated explants from the frontal cortex of three rhesus brains with medium alone or medium with added live Borrelia burgdorferi for 6, 12, and 24 h and isolated RNA from each group was used for RNA sequencing with further bioinformatic analysis. Transcriptomic differences between the ex vivo model of live Borrelia burgdorferi with rhesus frontal cortex tissue explants and the controls during the progression of the infection were identified. A total of 2249, 1064, and 420 genes were significantly altered, of which 80.7, 52.9, and 19.8% were upregulated and 19.3, 47.1, 80.2% were downregulated at 6, 12, and 24 h, respectively. Gene ontology and KEGG pathway analyses revealed various pathways related to immune and inflammatory responses during the spirochete infection were enriched which is suggested to have a causal role in the pathogenesis of neurological Lyme disease. Moreover, we propose that the overexpressed FOLR2 which was demonstrated by the real-time PCR and western blotting could play a key role in neuroinflammation of the neuroborreliosis based on PPI analysis for the first time. To our knowledge, this is the first study to provide comprehensive information regarding the transcriptomic signatures that occur in the frontal cortex of the brain upon exposure to Borrelia burgdorferi, and suggest that FOLR2 is a promising target that is associated with neuroinflammation and may represent a new diagnostic or therapeutic marker in LNB.
Collapse
Affiliation(s)
- Zhe Ding
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Mingbiao Ma
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lvyan Tao
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Yun Peng
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Yuanyuan Han
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Luyun Sun
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China
| | - Xiting Dai
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Zhenhua Ji
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Ruolan Bai
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Miaomiao Jian
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Taigui Chen
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lisha Luo
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Feng Wang
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China
| | - Yunfeng Bi
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China
| | - Aihua Liu
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China.,Yunnan Province Integrative Innovation Center for Public Health, Diseases Prevention and Control, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Fukai Bao
- Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities, Kunming Medical University, Kunming, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,Yunnan Province Integrative Innovation Center for Public Health, Diseases Prevention and Control, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| |
Collapse
|
26
|
Neuronal loss or dysfunction in patients with early Lyme neuroborreliosis: a proton magnetic resonance spectroscopy study of the brain. J Neurol 2019; 266:1937-1943. [PMID: 31076877 PMCID: PMC6647218 DOI: 10.1007/s00415-019-09359-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/08/2019] [Accepted: 05/03/2019] [Indexed: 11/22/2022]
Abstract
Background We hypothesized that since Borrelia burgdorferi causes systemic inflammation and infects the brain, it may lead to alterations in cerebral metabolism, as measured by 1H-magnetic resonance spectroscopy (1H-MRS). The purpose of our study was to determine whether 1H-MRS could detect brain metabolite alterations in patients with early Lyme neuroborreliosis (LNB) in normal-appearing brain tissue on the conventional magnetic resonance imaging (MRI). Methods Twenty-six patients diagnosed with early LNB and twenty-six healthy volunteers as a control group have been involved in the study. All of them underwent routine MRI protocol using 3.0-T MRI scanner. 1H-MRS examinations were performed with repetition time (TR) = 2000 ms, and echo time (TE) = 135 ms. Single voxels were positioned in the anterior and posterior parts of the right and left frontal lobes. Results We found a statistically significant decrease of the N-acetylaspartate/creatine ratio within the anterior part of the right and left frontal lobes (p ≤ 0.001 and p = 0.001 respectively) and in the posterior part of the right and left frontal lobes (p ≤ 0.001 and 0.031) in the patients with LNB. Conclusion A significant reduction in NAA/Cr ratio in comparison with the controls suggests the presence of diffuse neuronal loss in patients with early LNB.
Collapse
|
27
|
Scott JD, Clark KL, Durden LA. Presence of Babesia odocoilei and Borrelia burgdorferi Sensu Stricto in a Tick and Dual Parasitism of Amblyomma inornatum and Ixodes scapularis on a Bird in Canada. Healthcare (Basel) 2019; 7:healthcare7010046. [PMID: 30897803 PMCID: PMC6473902 DOI: 10.3390/healthcare7010046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
Wild birds transport ticks into Canada that harbor a diversity of zoonotic pathogens. However, medical practitioners often question how these zoonotic pathogens are present in their locality. In this study, we provide the first report of an Amblyomma inornatum tick cofeeding with a blacklegged tick, Ixodes scapularis, which parasitized a Veery, Catharus fuscescens—a neotropical songbird. Using the flagellin (flaB) gene of the Lyme disease bacterium, Borrelia burgdorferi sensu lato, and the 18S rRNA gene of the Babesia piroplasm, a malaria-like microorganism, we detected Borrelia burgdorferi sensu stricto and Babesia odocoilei, respectively, in an I. scapularis nymph. After the molt, these ticks can bite humans. Furthermore, this is the first documentation of B. odocoilei in a tick parasitizing a bird. Our findings substantiate the fact that migratory songbirds transport neotropical ticks long distances, and import them into Canada during northward spring migration. Health care practitioners need to be aware that migratory songbirds transport pathogen-laden ticks into Canada annually, and pose an unforeseen health risk to Canadians.
Collapse
Affiliation(s)
- John D Scott
- International Lyme and Associated Diseases Society, 2 Wisconsin Circle, Suite 700, Chevy Chase, MD 20185-7007, USA.
| | - Kerry L Clark
- Environmental Epidemiology Research Laboratory, Department of Public Health, University of North Florida, Jacksonville, FL 32224, USA.
| | - Lance A Durden
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA.
| |
Collapse
|
28
|
Jarosz AC, Badawi A. Metabolites of prostaglandin synthases as potential biomarkers of Lyme disease severity and symptom resolution. Inflamm Res 2018; 68:7-17. [PMID: 30121835 PMCID: PMC6314976 DOI: 10.1007/s00011-018-1180-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 01/09/2023] Open
Abstract
Background Lyme disease or Lyme borreliosis (LB) is the commonest vector-borne disease in the North America. It is an inflammatory disease caused by the bacterium Borrelia burgdorferi. The role of the inflammatory processes mediated by prostaglandins (PGs), thromboxanes and leukotrienes (LTs) in LB severity and symptoms resolution is yet to be elucidated. Objectives We aim to systematically review and evaluate the role of PGs and related lipid mediators in the induction and resolution of inflammation in LB. Methods We conducted a comprehensive search in PubMed, Ovid MEDLINE(R), Embase and Embase Classic to identify cell-culture, animal and human studies reporting the changes in PGs and related lipid mediators of inflammation during the course of LB. Results We identified 18 studies to be included into this systematic review. The selected reports consisted of seven cell-culture studies, seven animal studies, and four human studies (from three patient populations). Results from cell-culture and animal studies suggest that PGs and other lipid mediators of inflammation are elevated in LB and may contribute to disease development. The limited number of human studies showed that subjects with Lyme meningitis, Lyme arthritis (LA) and antibiotic-refractory LA had increased levels of an array of PGs and lipid mediators (e.g., LTB4, 8-isoPGF2α, and phospholipases A2 activity). Levels of these markers were significantly reduced following the treatment with antibiotics or non-steroidal anti-inflammatory drugs. Conclusion Dysregulation of prostaglandins and related lipid mediators may play a role in the etiology of LB and persistence of inflammation that may lead to long-term complications. Further investigation into the precise levels of a wide range of PGs and related factors is critical as it may propose novel markers that can be used for early diagnosis. Electronic supplementary material The online version of this article (10.1007/s00011-018-1180-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alicia Caroline Jarosz
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S3E2, Canada
| | - Alaa Badawi
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S3E2, Canada.
- Public Health Risk Sciences Division, Public Health Agency of Canada, 180 Queen Street West, Toronto, ON, M5V 3L7, Canada.
| |
Collapse
|
29
|
Scott JD, Clark KL, Foley JE, Bierman BC, Durden LA. Far-Reaching Dispersal of Borrelia burgdorferi Sensu Lato-Infected Blacklegged Ticks by Migratory Songbirds in Canada. Healthcare (Basel) 2018; 6:E89. [PMID: 30044388 PMCID: PMC6164468 DOI: 10.3390/healthcare6030089] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Lyme disease has been documented in northern areas of Canada, but the source of the etiological bacterium, Borrelia burgdorferi sensu lato (Bbsl) has been in doubt. We collected 87 ticks from 44 songbirds during 2017, and 24 (39%) of 62 nymphs of the blacklegged tick, Ixodes scapularis, were positive for Bbsl. We provide the first report of Bbsl-infected, songbird-transported I. scapularis in Cape Breton, Nova Scotia; Newfoundland and Labrador; north-central Manitoba, and Alberta. Notably, we report the northernmost account of Bbsl-infected ticks parasitizing a bird in Canada. DNA extraction, PCR amplification, and DNA sequencing reveal that these Bbsl amplicons belong to Borrelia burgdorferi sensu stricto (Bbss), which is pathogenic to humans. Based on our findings, health-care providers should be aware that migratory songbirds widely disperse B. burgdorferi-infected I. scapularis in Canada's North, and local residents do not have to visit an endemic area to contract Lyme disease.
Collapse
Affiliation(s)
- John D Scott
- International Lyme and Associated Diseases Society, Bethesda, MD 20827, USA.
| | - Kerry L Clark
- Epidemiology & Environmental Health, University of North Florida, Jacksonville, FL 32224, USA.
| | - Janet E Foley
- Vector-borne Disease Epidemiology and Department of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | - Bradley C Bierman
- Epidemiology & Environmental Health, University of North Florida, Jacksonville, FL 32224, USA.
| | - Lance A Durden
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA.
| |
Collapse
|
30
|
Parthasarathy G, Philipp MT. Intracellular TLR7 is activated in human oligodendrocytes in response to Borrelia burgdorferi exposure. Neurosci Lett 2018; 671:38-42. [PMID: 29408631 PMCID: PMC5889718 DOI: 10.1016/j.neulet.2018.01.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/05/2018] [Accepted: 01/31/2018] [Indexed: 01/06/2023]
Abstract
Lyme neuroborreliosis, caused by the gram-negative bacterium Borrelia burgdorferi, may affect the central and/or peripheral nervous systems. In previous studies, we showed that human oligodendrocytes exposed to the bacteria undergo apoptosis in an inflammatory environment, and that inflammatory pathways trigger cell-death pathways. We further demonstrated that several receptor tyrosine kinases were involved in triggering downstream effects, leading to inflammation and apoptosis. Toll-like receptors TLR2 and TLR5, which are commonly studied receptors in Lyme disease, only had a minimal role in inflammatory processes. To delineate the role of other TLRs, if any, real-time RT-PCR array experiments were carried out as an initial screen. Along with several inflammatory genes, TLR7 mRNA was upregulated in cells exposed to B. burgdorferi. Further analysis by immunohistochemistry showed that the TLR7 protein is present in readily detectable amounts, although no discernible differences could be seen between medium and B. burgdorferi-exposed cells by this technique. Nevertheless, use of specific inhibitors and siRNA showed that TLR7 is involved in inducing IL-6 and CCL2 in a dose dependent manner, and likely CXCL8. Triggering an intracellular receptor such as TLR7, which senses RNA, in typically non-phagocytic oligodendrocytes indicates either a niche for the bacterium inside the cell or novel uptake of nucleic acids to initiate inflammatory responses.
Collapse
Affiliation(s)
- Geetha Parthasarathy
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA.
| | - Mario T Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA.
| |
Collapse
|
31
|
Miklossy J. Bacterial Amyloid and DNA are Important Constituents of Senile Plaques: Further Evidence of the Spirochetal and Biofilm Nature of Senile Plaques. J Alzheimers Dis 2018; 53:1459-73. [PMID: 27314530 PMCID: PMC4981904 DOI: 10.3233/jad-160451] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
It has long been known that spirochetes form clumps or micro colonies in vitro and in vivo. Cortical spirochetal colonies in syphilitic dementia were considered as reproductive centers for spirochetes. Historic and recent data demonstrate that senile plaques in Alzheimer’s disease (AD) are made up by spirochetes. Spirochetes, are able to form biofilm in vitro. Senile plaques are also reported to contain elements of biofilm constituents. We expected that AβPP and Aβ (the main components of senile plaques) also occur in pure spirochetal biofilms, and bacterial DNA (an important component of biofilm) is also present in senile plaques. Histochemical, immunohistochemical, and in situ hybridization techniques and the TUNEL assay were used to answer these questions. The results obtained demonstrate that Aβ and DNA, including spirochete-specific DNA, are key components of both pure spirochetal biofilms and senile plaques in AD and confirm the biofilm nature of senile plaques. These results validate validate previous observations that AβPP and/or an AβPP-like amyloidogenic protein are an integral part of spirochetes, and indicate that bacterial and host derived Aβ are both constituents of senile plaques. DNA fragmentation in senile plaques further confirms their bacterial nature and provides biochemical evidence for spirochetal cell death. Spirochetes evade host defenses, locate intracellularly, form more resistant atypical forms and notably biofilms, which contribute to sustain chronic infection and inflammation and explain the slowly progressive course of dementia in AD. To consider co-infecting microorganisms is equally important, as multi-species biofilms result in a higher resistance to treatments and a more severe dementia.
Collapse
Affiliation(s)
- Judith Miklossy
- Correspondence to: Judith Miklossy, Prevention Alzheimer International Foundation, International Alzheimer Research Centre, Martigny-Croix, CP 16, 1921, Switzerland. Tel.: +41 79 207 4442/27 722 0652; E-mail:
| |
Collapse
|
32
|
Abstract
Inflammasomes are responsible for the maturation of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-18, and IL-33 and activation of inflammatory cell death, pyroptosis. They assemble in response to cellular infection and stress or to tissue damage, promote inflammatory reactions, and are important in regulating innate immunity particularly by acting as platforms for activation of caspase proteases. They appear to be involved in several pathological processes activated by microbes including Alzheimer's disease (AD). Best characterized in microbial pathogenesis is the nucleotide-binding domain and leucine-rich repeat (NLR)-protein 3 (NLRP3) inflammasome. AD is a neurodegenerative condition in which the neuropathological hallmarks are the deposition of amyloid-β (Aβ) and hyperphosphorylated tau protein coated neurofibrillary tangles. For decades, the role of the innate immune system in the etiology of AD was considered less important, but the recently discovered inflammatory genes by genome-wide association studies driving inflammation in this disease has changed this view. Innate immune inflammatory activity in the AD brain can result from the pathological hallmark protein Aβ as well as from specific bacterial infections that tend to possess weak immunostimulatory responses for peripheral blood myeloid cell recruitment to the brain. The weak immunostimulatory activity is a consequence of their immune evasion strategies and survival. In this review we discuss the possibility that inflammasomes, particularly via the NLR family of proteins NLRP3 are involved in the pathogenesis of AD. In addition, we discuss the plausible contribution of specific bacteria playing a role in influencing the activity of the NLRP3 inflammasome to AD progression.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Sim K Singhrao
- Oral & Dental Sciences Research Group, College of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| |
Collapse
|
33
|
Scannapieco FA, Cantos A. Oral inflammation and infection, and chronic medical diseases: implications for the elderly. Periodontol 2000 2018; 72:153-75. [PMID: 27501498 DOI: 10.1111/prd.12129] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2015] [Indexed: 12/12/2022]
Abstract
Oral diseases, such as caries and periodontitis, not only have local effects on the dentition and on tooth-supporting tissues but also may impact a number of systemic conditions. Emerging evidence suggests that poor oral health influences the initiation and/or progression of diseases such as atherosclerosis (with sequelae including myocardial infarction and stoke), diabetes mellitus and neurodegenerative diseases (such as Alzheimer's disease, rheumatoid arthritis and others). Aspiration of oropharyngeal (including periodontal) bacteria causes pneumonia, especially in hospitalized patients and the elderly, and may influence the course of chronic obstructive pulmonary disease. This article addresses several pertinent aspects related to the medical implications of periodontal disease in the elderly. There is moderate evidence that improved oral hygiene may help prevent aspiration pneumonia in high-risk patients. For other medical conditions, because of the absence of well-designed randomized clinical trials in elderly patients, no specific guidance can be provided regarding oral hygiene or periodontal interventions that enhance the medical management of older adults.
Collapse
|
34
|
Embers ME, Hasenkampf NR, Jacobs MB, Tardo AC, Doyle-Meyers LA, Philipp MT, Hodzic E. Variable manifestations, diverse seroreactivity and post-treatment persistence in non-human primates exposed to Borrelia burgdorferi by tick feeding. PLoS One 2017; 12:e0189071. [PMID: 29236732 PMCID: PMC5728523 DOI: 10.1371/journal.pone.0189071] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/17/2017] [Indexed: 01/21/2023] Open
Abstract
The efficacy and accepted regimen of antibiotic treatment for Lyme disease has been a point of significant contention among physicians and patients. While experimental studies in animals have offered evidence of post-treatment persistence of Borrelia burgdorferi, variations in methodology, detection methods and limitations of the models have led to some uncertainty with respect to translation of these results to human infection. With all stages of clinical Lyme disease having previously been described in nonhuman primates, this animal model was selected in order to most closely mimic human infection and response to treatment. Rhesus macaques were inoculated with B. burgdorferi by tick bite and a portion were treated with recommended doses of doxycycline for 28 days at four months post-inoculation. Signs of infection, clinical pathology, and antibody responses to a set of five antigens were monitored throughout the ~1.2 year study. Persistence of B. burgdorferi was evaluated using xenodiagnosis, bioassays in mice, multiple methods of molecular detection, immunostaining with polyclonal and monoclonal antibodies and an in vivo culture system. Our results demonstrate host-dependent signs of infection and variation in antibody responses. In addition, we observed evidence of persistent, intact, metabolically-active B. burgdorferi after antibiotic treatment of disseminated infection and showed that persistence may not be reflected by maintenance of specific antibody production by the host.
Collapse
Affiliation(s)
- Monica E. Embers
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
- * E-mail:
| | - Nicole R. Hasenkampf
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Mary B. Jacobs
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Amanda C. Tardo
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Lara A. Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Mario T. Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Emir Hodzic
- Center for Comparative Medicine, Schools of Medicine and Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| |
Collapse
|
35
|
Burmeister AR, Johnson MB, Chauhan VS, Moerdyk-Schauwecker MJ, Young AD, Cooley ID, Martinez AN, Ramesh G, Philipp MT, Marriott I. Human microglia and astrocytes constitutively express the neurokinin-1 receptor and functionally respond to substance P. J Neuroinflammation 2017; 14:245. [PMID: 29237453 PMCID: PMC5729418 DOI: 10.1186/s12974-017-1012-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022] Open
Abstract
Background The tachykinin substance P (SP) is recognized to exacerbate inflammation at peripheral sites via its target receptor, neurokinin 1 receptor (NK-1R), expressed by leukocytes. More recently, SP/NK-1R interactions have been associated with severe neuroinflammation and neuronal damage. We have previously demonstrated that NK-1R antagonists can limit neuroinflammatory damage in a mouse model of bacterial meningitis. Furthermore, we have since shown that these agents can attenuate bacteria-induced neuronal and glial inflammatory mediator production in nonhuman primate (NHP) brain explants and isolated neuronal cells, and following in vivo infection. Methods In the present study, we have assessed the ability of NHP brain explants, primary human microglia and astrocytes, and immortalized human glial cell lines to express NK-1R isoforms. We have utilized RT-PCR, immunoblot analysis, immunofluorescent microscopy, and/or flow cytometric analysis, to quantify NK-1R expression in each, at rest, or following bacterial challenge. Furthermore, we have assessed the ability of human microglia to respond to SP by immunoblot analysis of NF-kB nuclear translocation and determined the ability of this neuropeptide to augment inflammatory cytokine release and neurotoxic mediator production by human astrocytes using an ELISA and a neuronal cell toxicity assay, respectively. Results We demonstrate that human microglial and astrocytic cells as well as NHP brain tissue constitutively express robust levels of the full-length NK-1R isoform. In addition, we demonstrate that the expression of NK-1R by human astrocytes can be further elevated following exposure to disparate bacterial pathogens or their components. Importantly, we have demonstrated that NK-1R is functional in both human microglia and astrocytes and show that SP can augment the inflammatory and/or neurotoxic immune responses of glial cells to disparate and clinically relevant bacterial pathogens. Conclusions The robust constitutive and functional expression of the full-length NK-1R isoform by human microglia and astrocytes, and the ability of SP to augment inflammatory signaling pathways and mediator production by these cells, support the contention that SP/NK-1R interactions play a significant role in the damaging neuroinflammation associated with conditions such as bacterial meningitis.
Collapse
Affiliation(s)
- Amanda R Burmeister
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - M Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - Vinita S Chauhan
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - Megan J Moerdyk-Schauwecker
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - Ada D Young
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - Ian D Cooley
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA
| | - Alejandra N Martinez
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Geeta Ramesh
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Mario T Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, 28223, USA.
| |
Collapse
|
36
|
Crossland NA, Alvarez X, Embers ME. Late Disseminated Lyme Disease: Associated Pathology and Spirochete Persistence Posttreatment in Rhesus Macaques. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:672-682. [PMID: 29242055 PMCID: PMC5840488 DOI: 10.1016/j.ajpath.2017.11.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
Abstract
Nonhuman primates currently serve as the best experimental model for Lyme disease because of their close genetic homology with humans and demonstration of all three phases of disease after infection with Borrelia burgdorferi. We investigated the pathology associated with late disseminated Lyme disease (12 to 13 months after tick inoculation) in doxycycline-treated (28 days; 5 mg/kg, oral, twice daily) and untreated rhesus macaques. Minimal to moderate lymphoplasmacytic inflammation, with a predilection for perivascular spaces and collagenous tissues, was observed in multiple tissues, including the cerebral leptomeninges, brainstem, peripheral nerves from both fore and hind limbs, stifle synovium and perisynovial adipose tissue, urinary bladder, skeletal muscle, myocardium, and visceral pericardium. Indirect immunofluorescence assays that combined monoclonal (outer surface protein A) and polyclonal antibodies were performed on all tissue sections that contained inflammation. Rare morphologically intact spirochetes were observed in the brains of two treated rhesus macaques, the heart of one treated rhesus macaque, and adjacent to a peripheral nerve of an untreated animal. Borrelia antigen staining of probable spirochete cross sections was also observed in heart, skeletal muscle, and near peripheral nerves of treated and untreated animals. These findings support the notion that chronic Lyme disease symptoms can be attributable to residual inflammation in and around tissues that harbor a low burden of persistent host-adapted spirochetes and/or residual antigen.
Collapse
Affiliation(s)
- Nicholas A Crossland
- Division of Bacteriology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, Louisiana; Division of Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, Louisiana
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, Louisiana
| | - Monica E Embers
- Division of Bacteriology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, Louisiana; Division of Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, Louisiana.
| |
Collapse
|
37
|
Yang J, Han X, Liu A, Bao F, Peng Y, Tao L, Ma M, Bai R, Dai X. Chemokine CXC Ligand 13 in Cerebrospinal Fluid Can Be Used as an Early Diagnostic Biomarker for Lyme Neuroborreliosis: A Meta-Analysis. J Interferon Cytokine Res 2017; 37:433-439. [PMID: 28972436 DOI: 10.1089/jir.2016.0101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Lyme neuroborreliosis (LNB), which is the most common neurological manifestation of Lyme disease (LD), seriously impairs both the central and peripheral nervous systems. Current LNB diagnostic methods and criteria are not very effective. Recently, several studies have indicated that a high concentration of the chemokine CXC ligand 13 (CXCL13) in cerebrospinal fluid (CSF) could be used as a new biomarker for the diagnosis of LNB. Thus, we carried out a meta-analysis to systematically analyze the data from these studies to evaluate the value of CXCL13 as an LNB biomarker. After searching for articles in several databases, including PubMed, Embase, the Cochrane Library, and the China National Knowledge Infrastructure (CNKI), we included 7 articles in the meta-analysis with a total of 1299 patients with LNB or other neuroinflammatory diseases. From these 1299 patients, 343 patients with LNB served as the experimental group and 956 patients with other neuroinflammatory diseases or healthy individuals served as the control group. The analyses were performed using Meta-Disc1.4 statistical software. Based on the pooled specificity, sensitivity, positive likelihood ratio, negative likelihood ratio, diagnostic odds ratio, and summary receiver operating characteristic curve, we found that CXCL13 indeed has a high sensitivity and specificity for diagnosing LNB, which means that it can be used as a new diagnostic biomarker for the diagnosis of LNB.
Collapse
Affiliation(s)
- Jiaru Yang
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,2 Department of Biochemistry and Molecular Biology, Kunming Medical University , Kunming, China
| | - Xinlin Han
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,3 Department of Microbiology and Immunology, Kunming Medical University , Kunming, China
| | - Aihua Liu
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,2 Department of Biochemistry and Molecular Biology, Kunming Medical University , Kunming, China .,4 Institute for Tropical Medicine, Kunming Medical University , Kunming, China .,5 Yunnan Province Integrative Innovation Center for Public Health, Disease Prevention and Control, Kunming Medical University , Kunming, China .,6 Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases , Kunming, China
| | - Fukai Bao
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,3 Department of Microbiology and Immunology, Kunming Medical University , Kunming, China .,4 Institute for Tropical Medicine, Kunming Medical University , Kunming, China .,5 Yunnan Province Integrative Innovation Center for Public Health, Disease Prevention and Control, Kunming Medical University , Kunming, China .,6 Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases , Kunming, China
| | - Yun Peng
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,3 Department of Microbiology and Immunology, Kunming Medical University , Kunming, China
| | - Lüyan Tao
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,2 Department of Biochemistry and Molecular Biology, Kunming Medical University , Kunming, China
| | - Mingbiao Ma
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,3 Department of Microbiology and Immunology, Kunming Medical University , Kunming, China
| | - Ruolan Bai
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,2 Department of Biochemistry and Molecular Biology, Kunming Medical University , Kunming, China
| | - Xiting Dai
- 1 Yunnan Province Key Laboratory for Tropical Infectious Diseases in Universities , Kunming, China .,3 Department of Microbiology and Immunology, Kunming Medical University , Kunming, China
| |
Collapse
|
38
|
Abstract
Lyme disease is caused by the bacterium Borrelia burgdorferi and is transmitted to humans through the bite of infected blacklegged ticks. According to the Centers for Disease Control and Prevention, it is the most commonly reported vector-borne illness and the fifth most common disease in the National Notifiable Diseases Surveillance System. If left untreated, infection disseminates to the nervous system. The nonhuman primate model of Lyme disease of the nervous system, or Lyme neuroborreliosis, accurately mimics the aspects of the human illness. There is general recognition for the potential of infectious-related autoimmune inflammatory processes contributing to disease progression and clinical manifestations.
Collapse
Affiliation(s)
- David S Younger
- Division of Neuroepidemiology, Department of Neurology, New York University School of Medicine, New York, NY, USA; College of Global Public Health, New York University, New York, NY, USA.
| |
Collapse
|
39
|
Parthasarathy G, Philipp MT. Receptor tyrosine kinases play a significant role in human oligodendrocyte inflammation and cell death associated with the Lyme disease bacterium Borrelia burgdorferi. J Neuroinflammation 2017; 14:110. [PMID: 28558791 PMCID: PMC5450372 DOI: 10.1186/s12974-017-0883-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/18/2017] [Indexed: 12/28/2022] Open
Abstract
Background In previous studies, human oligodendrocytes were demonstrated to undergo apoptosis in the presence of Borrelia burgdorferi under an inflammatory milieu. Subsequently, we determined that the MEK/ERK pathway played a significant role in triggering downstream inflammation as well as apoptosis. However, the identity of receptors triggered by exposure to B. burgdorferi and initiating signaling events was unknown. Methods In this study, we explored the role of several TLR and EGFR/FGFR/PDGFR tyrosine kinase pathways in inducing inflammation in the presence of B. burgdorferi, using siRNA and/or inhibitors, in MO3.13 human oligodendrocytes. Cell death and apoptosis assays were also carried out in the presence or absence of specific receptor inhibitors along with the bacteria to determine the role of these receptors in apoptosis induction. The expression pattern of specific receptors with or without B. burgdorferi was also determined. Results TLRs 2 and 5 had a minimal role in inducing inflammation, particularly IL-6 production. Rather, their effect was mostly inhibitory, with TLR2 downregulation significantly upregulating CXCL8, and CXCL (1,2,3) levels, and TLR5 likely having a similar role in CXCL8, CXCL(1,2,3), and CCL5 levels. TLR4 contributed mostly towards CCL5 production. On the other hand, inhibition of all three EGF/FGF/PDGF receptors significantly downregulated all five of the inflammatory mediators tested even in the presence of B. burgdorferi. Their inhibition also downregulated overall cell death and apoptosis levels. The expression pattern of these receptors, as assessed by immunohistochemistry indicated that the PDGFRβ receptor was the most predominantly expressed receptor, followed by FGFR, although no significant differences were discernible between presence and absence of bacteria. Interestingly, inhibition of individual EGFR, FGFR, or PDGFR receptors did not indicate an individual role for any of these receptors in the overall downregulation of pathogenesis. Contrarily, suppression of FGFR signaling alone in the presence of bacteria significantly upregulated inflammatory mediator levels indicating that it might control an inhibitory pathway when triggered individually. Conclusions Unlike TLRs, EGF/FGF/PDGF receptors collectively play a significant role in the inflammation and apoptosis of human oligodendrocytes as mediated by B. burgdorferi. It is likely that these three receptors need to be triggered simultaneously to achieve this effect.
Collapse
Affiliation(s)
- Geetha Parthasarathy
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA
| | - Mario T Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703, Three Rivers Road, Covington, LA, 70433, USA.
| |
Collapse
|
40
|
Ramesh G, Martinez AN, Martin DS, Philipp MT. Effects of dexamethasone and meloxicam on Borrelia burgdorferi-induced inflammation in glial and neuronal cells of the central nervous system. J Neuroinflammation 2017; 14:28. [PMID: 28153013 PMCID: PMC5290621 DOI: 10.1186/s12974-017-0806-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/26/2017] [Indexed: 11/21/2022] Open
Abstract
Background Lyme neuroborreliosis (LNB), caused by the spirochete Borrelia burgdorferi (Bb), affects both the central and peripheral nervous systems. Previously, we reported that in a model of acute LNB in rhesus monkeys, treatment with the anti-inflammatory drug dexamethasone significantly reduced both pleocytosis and levels of cerebrospinal fluid (CSF) immune mediators that were induced by Bb. Dexamethasone also inhibited the formation of inflammatory, neurodegenerative, and demyelinating lesions in the brain and spinal cord of these animals. In contrast, these signs were evident in the infected animals that were left untreated or in those that were treated with meloxicam, a non-steroidal anti-inflammatory drug. Methods To address the differential anti-inflammatory effects of dexamethasone and meloxicam in the central nervous system (CNS), we evaluated the potential of these drugs to alter the levels of Bb-induced inflammatory mediators in culture supernatants of rhesus frontal cortex (FC) explants, primary rhesus astrocytes and microglia, and human oligodendrocytes. We also ascertained the potential of dexamethasone to modulate Bb-induced apoptosis in rhesus FC explants. As meloxicam is a known COX-2 inhibitor, we evaluated whether meloxicam altered the levels of COX-2 as induced by live Bb in cell lysates of primary rhesus astrocytes and microglia. Results Dexamethasone but not meloxicam significantly reduced the levels of several Bb-induced immune mediators in culture supernatants of FC explants, astrocytes, microglia, and oligodendrocytes. Dexamethasone also had a protective effect on Bb-induced neuronal and oligodendrocyte apoptosis in rhesus FC explants. Further, meloxicam significantly reduced the levels of Bb-induced COX-2 in microglia, while both Bb and meloxicam were unable to alter the constitutive levels of COX-2 in astrocytes. Conclusions These data indicate that dexamethasone and meloxicam have differential anti-inflammatory effects on Bb-induced inflammation in glial and neuronal cells of the CNS and help explain the in vivo findings of significantly reduced inflammatory mediators in the CSF and lack of inflammatory neurodegenerative lesions in the brain and spinal cord of Bb-infected animals that were treated with dexamethasone but not meloxicam. Signaling cascades altered by dexamethasone could serve as possible therapeutic targets for limiting CNS inflammation and tissue damage in LNB.
Collapse
Affiliation(s)
- Geeta Ramesh
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Alejandra N Martinez
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Dale S Martin
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA
| | - Mario T Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA. .,Department of Microbiology and Immunology, Tulane University Medical School, New Orleans, LA, USA.
| |
Collapse
|
41
|
Casselli T, Qureshi H, Peterson E, Perley D, Blake E, Jokinen B, Abbas A, Nechaev S, Watt JA, Dhasarathy A, Brissette CA. MicroRNA and mRNA Transcriptome Profiling in Primary Human Astrocytes Infected with Borrelia burgdorferi. PLoS One 2017; 12:e0170961. [PMID: 28135303 PMCID: PMC5279786 DOI: 10.1371/journal.pone.0170961] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/14/2017] [Indexed: 02/07/2023] Open
Abstract
Lyme disease is caused by infection with the bacterium Borrelia burgdorferi (Bb), which is transmitted to humans by deer ticks. The infection manifests usually as a rash and minor systemic symptoms; however, the bacteria can spread to other tissues, causing joint pain, carditis, and neurological symptoms. Lyme neuroborreliosis presents itself in several ways, such as Bell's palsy, meningitis, and encephalitis. The molecular basis for neuroborreliosis is poorly understood. Analysis of the changes in the expression levels of messenger RNAs and non-coding RNAs, including microRNAs, following Bb infection could therefore provide vital information on the pathogenesis and clinical symptoms of neuroborreliosis. To this end, we used cultured primary human astrocytes, key responders to CNS infection and important components of the blood-brain barrier, as a model system to study RNA and microRNA changes in the CNS caused by Bb. Using whole transcriptome RNA-seq, we found significant changes in 38 microRNAs and 275 mRNAs at 24 and 48 hours following Bb infection. Several of the RNA changes affect pathways involved in immune response, development, chromatin assembly (including histones) and cell adhesion. Further, several of the microRNA predicted target mRNAs were also differentially regulated. Overall, our results indicate that exposure to Bb causes significant changes to the transcriptome and microRNA profile of astrocytes, which has implications in the pathogenesis, and hence potential treatment strategies to combat this disease.
Collapse
Affiliation(s)
- Timothy Casselli
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Humaira Qureshi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Elizabeth Peterson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Emily Blake
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Bradley Jokinen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Ata Abbas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Sergei Nechaev
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - John A. Watt
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Catherine A. Brissette
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| |
Collapse
|
42
|
The Lyme Disease Pathogen Borrelia burgdorferi Infects Murine Bone and Induces Trabecular Bone Loss. Infect Immun 2017; 85:IAI.00781-16. [PMID: 27956598 PMCID: PMC5278181 DOI: 10.1128/iai.00781-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023] Open
Abstract
Lyme disease is caused by members of the Borrelia burgdorferisensu lato species complex. Arthritis is a well-known late-stage pathology of Lyme disease, but the effects of B. burgdorferi infection on bone at sites other than articular surfaces are largely unknown. In this study, we investigated whether B. burgdorferi infection affects bone health in mice. In mice inoculated with B. burgdorferi or vehicle (mock infection), we measured the presence of B. burgdorferi DNA in bones, bone mineral density (BMD), bone formation rates, biomechanical properties, cellular composition, and two- and three-dimensional features of bone microarchitecture. B. burgdorferi DNA was detected in bone. In the long bones, increasing B. burgdorferi DNA copy number correlated with reductions in areal and trabecular volumetric BMDs. Trabecular regions of femora exhibited significant, copy number-correlated microarchitectural disruption, but BMD, microarchitectural, and biomechanical properties of cortical bone were not affected. Bone loss in tibiae was not due to increased osteoclast numbers or bone-resorbing surface area, but it was associated with reduced osteoblast numbers, implying that bone loss in long bones was due to impaired bone building. Osteoid-producing and mineralization activities of existing osteoblasts were unaffected by infection. Therefore, deterioration of trabecular bone was not dependent on inhibition of osteoblast function but was more likely caused by blockade of osteoblastogenesis, reduced osteoblast survival, and/or induction of osteoblast death. Together, these data represent the first evidence that B. burgdorferi infection induces bone loss in mice and suggest that this phenotype results from inhibition of bone building rather than increased bone resorption.
Collapse
|
43
|
Scott JD, Foley JE, Anderson JF, Clark KL, Durden LA. Detection of Lyme Disease Bacterium, Borrelia burgdorferi sensu lato, in Blacklegged Ticks Collected in the Grand River Valley, Ontario, Canada. Int J Med Sci 2017; 14:150-158. [PMID: 28260991 PMCID: PMC5332844 DOI: 10.7150/ijms.17763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/28/2016] [Indexed: 01/08/2023] Open
Abstract
We document the presence of blacklegged ticks, Ixodes scapularis, in the Grand River valley, Centre Wellington, Ontario. Overall, 15 (36%) of 42 I. scapularis adults collected from 41 mammalian hosts (dogs, cats, humans) were positive for the Lyme disease bacterium, Borrelia burgdorferi sensu lato (s.l.). Using real-time PCR testing and DNA sequencing of the flagellin (fla) gene, we determined that Borrelia amplicons extracted from I. scapularis adults belonged to B. burgdorferi sensu stricto (s.s.), which is pathogenic to humans and certain domestic animals. Based on the distribution of I. scapularis adults within the river basin, it appears likely that migratory birds provide an annual influx of I. scapularis immatures during northward spring migration. Health-care providers need to be aware that local residents can present with Lyme disease symptoms anytime during the year.
Collapse
Affiliation(s)
- John D Scott
- Research Division, Lyme Ontario, Fergus, Ontario Canada N1M 2L7
| | - Janet E Foley
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, USA 95616
| | - John F Anderson
- Department of Entomology and Center for Vector Ecology and Zoonotic Diseases. The Connecticut Agricultural Experiment Station, New Haven, Connecticut, USA 06504
| | - Kerry L Clark
- Epidemiology & Environmental Health, Department of Public Health, University of North Florida, Jacksonville, USA 32224
| | - Lance A Durden
- Department of Biology, Georgia Southern University, Statesboro, Georgia 30458, USA
| |
Collapse
|
44
|
Kovalchuka L, Cvetkova S, Trofimova J, Eglite J, Gintere S, Lucenko I, Oczko-Grzesik B, Viksna L, Krumina A. Immunogenetic Markers Definition in Latvian Patients with Lyme Borreliosis and Lyme Neuroborreliosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13121194. [PMID: 27916969 PMCID: PMC5201335 DOI: 10.3390/ijerph13121194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 12/29/2022]
Abstract
The aim of this study was to determine the human leukocyte antigen (HLA)-DRB1 alleles in two groups of patients in Latvia: patients with Lyme borreliosis and patients with Lyme neuroborreliosis. The study included 216 patients with Lyme borreliosis, 29 patients with Lyme neuroborreliosis and 282 control persons. All surveyed persons were residents of Latvia. The HLA-DR genotyping was performed by polymerase chain reaction- sequence specific primer (PCR-SSP). The predisposition to the Lyme borreliosis is associated with the HLA-DRB1*07, -DRB1*17(03), -DRB1*04, -DRB1*15(02) alleles. The allele -DRB1*11(05), -DRB1*14(06) and -DRB1*13(06) were significantly more frequent in controls. In-group with Lyme neuroborreliosis differences were found for the -DRB1*07 and -DRB1*04 alleles, but only HLA-DRB1*07 allele was statistically significant after Bonferroni correction and associated with Lyme neuroborreliosis in Latvian patients.
Collapse
Affiliation(s)
- Lilija Kovalchuka
- Institute of Food Safety, Animal Health and Environment BIOR, Riga LV-1076, Latvia.
| | - Svetlana Cvetkova
- Institute of Food Safety, Animal Health and Environment BIOR, Riga LV-1076, Latvia.
| | - Julija Trofimova
- Institute of Food Safety, Animal Health and Environment BIOR, Riga LV-1076, Latvia.
| | - Jelena Eglite
- Laboratory of Clinical Immunology and Immunogenetic, Riga Stradiņš University, Riga LV-1067, Latvia.
| | - Sandra Gintere
- Department of Family Medicine, Riga Stradiņš University, Riga LV-1067, Latvia.
| | - Irina Lucenko
- Centre for Disease Prevention and Control of Latvia, Riga LV-1005, Latvia.
| | - Barbara Oczko-Grzesik
- Department of Infectious Diseases, Medical University of Silesia, 40-055 Katowice, Poland.
| | - Ludmila Viksna
- Department of Infectology and Dermatology, Riga Stradiņš University, Riga LV-1006, Latvia.
| | - Angelika Krumina
- Institute of Food Safety, Animal Health and Environment BIOR, Riga LV-1076, Latvia.
- Department of Infectology and Dermatology, Riga Stradiņš University, Riga LV-1006, Latvia.
| |
Collapse
|
45
|
Abstract
The chemokine, C-X-C motif ligand 13 (CXCL13), is constitutively expressed in lymphoid organs and controls the recruitment and compartmentalization of lymphocytes and antigen presenting cells within these specialized structures. Recent data, however, also show induction of this molecule under a variety of circumstances during central nervous system (CNS) inflammation. While its role(s) in the pathogenesis of neoplastic, infectious and autoimmune disorders of the CNS remain incompletely understood, growing evidence suggests that CXCL13 could become a relevant therapeutic target in at least some of these conditions. This review focuses on the diseases, cellular sources and external factors known to regulate CXCL13 production in the inflamed CNS.
Collapse
Affiliation(s)
- David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
46
|
Harris SA, Harris EA. Herpes Simplex Virus Type 1 and Other Pathogens are Key Causative Factors in Sporadic Alzheimer's Disease. J Alzheimers Dis 2016; 48:319-53. [PMID: 26401998 PMCID: PMC4923765 DOI: 10.3233/jad-142853] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review focuses on research in epidemiology, neuropathology, molecular biology, and genetics regarding the hypothesis that pathogens interact with susceptibility genes and are causative in sporadic Alzheimer's disease (AD). Sporadic AD is a complex multifactorial neurodegenerative disease with evidence indicating coexisting multi-pathogen and inflammatory etiologies. There are significant associations between AD and various pathogens, including Herpes simplex virus type 1 (HSV-1), Cytomegalovirus, and other Herpesviridae, Chlamydophila pneumoniae, spirochetes, Helicobacter pylori, and various periodontal pathogens. These pathogens are able to evade destruction by the host immune system, leading to persistent infection. Bacterial and viral DNA and RNA and bacterial ligands increase the expression of pro-inflammatory molecules and activate the innate and adaptive immune systems. Evidence demonstrates that pathogens directly and indirectly induce AD pathology, including amyloid-β (Aβ) accumulation, phosphorylation of tau protein, neuronal injury, and apoptosis. Chronic brain infection with HSV-1, Chlamydophila pneumoniae, and spirochetes results in complex processes that interact to cause a vicious cycle of uncontrolled neuroinflammation and neurodegeneration. Infections such as Cytomegalovirus, Helicobacter pylori, and periodontal pathogens induce production of systemic pro-inflammatory cytokines that may cross the blood-brain barrier to promote neurodegeneration. Pathogen-induced inflammation and central nervous system accumulation of Aβ damages the blood-brain barrier, which contributes to the pathophysiology of AD. Apolipoprotein E4 (ApoE4) enhances brain infiltration by pathogens including HSV-1 and Chlamydophila pneumoniae. ApoE4 is also associated with an increased pro-inflammatory response by the immune system. Potential antimicrobial treatments for AD are discussed, including the rationale for antiviral and antibiotic clinical trials.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
47
|
Johnstone LK, Engiles JB, Aceto H, Buechner-Maxwell V, Divers T, Gardner R, Levine R, Scherrer N, Tewari D, Tomlinson J, Johnson AL. Retrospective Evaluation of Horses Diagnosed with Neuroborreliosis on Postmortem Examination: 16 Cases (2004-2015). J Vet Intern Med 2016; 30:1305-12. [PMID: 27327172 PMCID: PMC5094551 DOI: 10.1111/jvim.14369] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/30/2016] [Accepted: 05/26/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Equine neuroborreliosis (NB), Lyme disease, is difficult to diagnose and has limited description in the literature. OBJECTIVE Provide a detailed description of clinical signs, diagnostic, and pathologic findings of horses with NB. ANIMALS Sixteen horses with histologically confirmed NB. METHODS Retrospective review of medical records at the University of Pennsylvania and via an ACVIM listserv query with inclusion criteria requiring possible exposure to Borrelia burgdorferi and histologic findings consistent with previous reports of NB without evidence of other disease. RESULTS Sixteen horses were identified, 12 of which had additional evidence of NB. Clinical signs were variable including muscle atrophy or weight loss (12), cranial nerve deficits (11), ataxia (10), changes in behavior (9), dysphagia (7), fasciculations (6), neck stiffness (6), episodic respiratory distress (5), uveitis (5), fever (2), joint effusion (2), and cardiac arrhythmias (1). Serologic analysis was positive for B. burgdorferi infection in 6/13 cases tested. CSF abnormalities were present in 8/13 cases tested, including xanthochromia (4/13), increased total protein (5/13; median: 91 mg/dL, range: 25-219 mg/dL), and a neutrophilic (6/13) or lymphocytic (2/13) pleocytosis (median: 25 nucleated cells/μL, range: 0-922 nucleated cells/μL). PCR on CSF for B. burgdorferi was negative in the 7 cases that were tested. CONCLUSION AND CLINICAL IMPORTANCE Diagnosis of equine NB is challenging due to variable clinical presentation and lack of sensitive and specific diagnostic tests. Negative serology and normal CSF analysis do not exclude the diagnosis of NB.
Collapse
Affiliation(s)
- L K Johnstone
- University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA
| | - J B Engiles
- University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA
| | - H Aceto
- University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA
| | - V Buechner-Maxwell
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA
| | - T Divers
- Cornell University College of Veterinary Medicine, Ithaca, NY
| | - R Gardner
- B.W. Furlong & Associates, Oldwick, NJ
| | - R Levine
- Henderson Veterinary Associates, Elizabethtown, PA
| | - N Scherrer
- University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA
| | - D Tewari
- Pennsylvania Veterinary Laboratory, Pennsylvania Department of Agriculture, Harrisburg, PA
| | - J Tomlinson
- Cornell University College of Veterinary Medicine, Ithaca, NY
| | - A L Johnson
- University of Pennsylvania School of Veterinary Medicine, New Bolton Center, Kennett Square, PA
| |
Collapse
|
48
|
Miraglia MC, Costa Franco MM, Rodriguez AM, Bellozi PMQ, Ferrari CC, Farias MI, Dennis VA, Barrionuevo P, de Oliveira ACP, Pitossi F, Kim KS, Delpino MV, Oliveira SC, Giambartolomei GH. Glial Cell-Elicited Activation of Brain Microvasculature in Response to Brucella abortus Infection Requires ASC Inflammasome-Dependent IL-1β Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:3794-3805. [PMID: 26983788 DOI: 10.4049/jimmunol.1500908] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 02/18/2016] [Indexed: 01/18/2023]
Abstract
Blood-brain barrier activation and/or dysfunction are a common feature of human neurobrucellosis, but the underlying pathogenic mechanisms are largely unknown. In this article, we describe an immune mechanism for inflammatory activation of human brain microvascular endothelial cells (HBMEC) in response to infection with Brucella abortus Infection of HBMEC with B. abortus induced the secretion of IL-6, IL-8, and MCP-1, and the upregulation of CD54 (ICAM-1), consistent with a state of activation. Culture supernatants (CS) from glial cells (astrocytes and microglia) infected with B. abortus also induced activation of HBMEC, but to a greater extent. Although B. abortus-infected glial cells secreted IL-1β and TNF-α, activation of HBMEC was dependent on IL-1β because CS from B. abortus-infected astrocytes and microglia deficient in caspase-1 and apoptosis-associated speck-like protein containing a CARD failed to induce HBMEC activation. Consistently, treatment of CS with neutralizing anti-IL-1β inhibited HBMEC activation. Both absent in melanoma 2 and Nod-like receptor containing a pyrin domain 3 are partially required for caspase-1 activation and IL-1β secretion, suggesting that multiple apoptosis-associated speck-like protein containing CARD-dependent inflammasomes contribute to IL-1β-induced activation of the brain microvasculature. Inflammasome-mediated IL-1β secretion in glial cells depends on TLR2 and MyD88 adapter-like/TIRAP. Finally, neutrophil and monocyte migration across HBMEC monolayers was increased by CS from Brucella-infected glial cells in an IL-1β-dependent fashion, and the infiltration of neutrophils into the brain parenchyma upon intracranial injection of B. abortus was diminished in the absence of Nod-like receptor containing a pyrin domain 3 and absent in melanoma 2. Our results indicate that innate immunity of the CNS set in motion by B. abortus contributes to the activation of the blood-brain barrier in neurobrucellosis and IL-1β mediates this phenomenon.
Collapse
Affiliation(s)
- M Cruz Miraglia
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Miriam M Costa Franco
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Ana M Rodriguez
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Paula M Q Bellozi
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Carina C Ferrari
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Maria I Farias
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104; and
| | - Paula Barrionuevo
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Antonio C P de Oliveira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Fernando Pitossi
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - M Victoria Delpino
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Guillermo H Giambartolomei
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina;
| |
Collapse
|
49
|
Jiang BC, Cao DL, Zhang X, Zhang ZJ, He LN, Li CH, Zhang WW, Wu XB, Berta T, Ji RR, Gao YJ. CXCL13 drives spinal astrocyte activation and neuropathic pain via CXCR5. J Clin Invest 2016; 126:745-61. [PMID: 26752644 DOI: 10.1172/jci81950] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Recent studies have implicated chemokines in microglial activation and pathogenesis of neuropathic pain. C-X-C motif chemokine 13 (CXCL13) is a B lymphocyte chemoattractant that activates CXCR5. Using the spinal nerve ligation (SNL) model of neuropathic pain, we found that CXCL13 was persistently upregulated in spinal cord neurons after SNL, resulting in spinal astrocyte activation via CXCR5 in mice. shRNA-mediated inhibition of CXCL13 in the spinal cord persistently attenuated SNL-induced neuropathic pain. Interestingly, CXCL13 expression was suppressed by miR-186-5p, a microRNA that colocalized with CXCL13 and was downregulated after SNL. Spinal overexpression of miR-186-5p decreased CXCL13 expression, alleviating neuropathic pain. Furthermore, SNL induced CXCR5 expression in spinal astrocytes, and neuropathic pain was abrogated in Cxcr5-/- mice. CXCR5 expression induced by SNL was required for the SNL-induced activation of spinal astrocytes and microglia. Intrathecal injection of CXCL13 was sufficient to induce pain hypersensitivity and astrocyte activation via CXCR5 and ERK. Finally, intrathecal injection of CXCL13-activated astrocytes induced mechanical allodynia in naive mice. Collectively, our findings reveal a neuronal/astrocytic interaction in the spinal cord by which neuronally produced CXCL13 activates astrocytes via CXCR5 to facilitate neuropathic pain. Thus, miR-186-5p and CXCL13/CXCR5-mediated astrocyte signaling may be suitable therapeutic targets for neuropathic pain.
Collapse
|
50
|
Scott JD, Foley JE, Clark KL, Anderson JF, Durden LA, Manord JM, Smith ML. Established Population of Blacklegged Ticks with High Infection Prevalence for the Lyme Disease Bacterium, Borrelia burgdorferi Sensu Lato, on Corkscrew Island, Kenora District, Ontario. Int J Med Sci 2016; 13:881-891. [PMID: 27877080 PMCID: PMC5118759 DOI: 10.7150/ijms.16922] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/26/2016] [Indexed: 12/24/2022] Open
Abstract
We document an established population of blacklegged ticks, Ixodes scapularis, on Corkscrew Island, Kenora District, Ontario, Canada. Primers of the outer surface protein A (OspA) gene, the flagellin (fla) gene, and the flagellin B (flaB) gene were used in the PCR assays to detect Borrelia burgdorferi sensu lato (s.l.), the Lyme disease bacterium. In all, 60 (73%) of 82 adult I. scapularis, were infected with B. burgdorferi s.l. As well, 6 (43%) of 14 unfed I. scapularis nymphs were positive for B. burgdorferi s.l. An I. scapularis larva was also collected from a deer mouse, and several unfed larvae were gathered by flagging leaf litter. Based on DNA sequencing of randomly selected Borrelia amplicons from six nymphal and adult I. scapularis ticks, primers for the flagellin (fla) and flagellin B (flaB) genes reveal the presence of B. burgdorferi sensu stricto (s.s.), a genospecies pathogenic to humans and certain domestic animals. We collected all 3 host-feeding life stages of I. scapularis in a single year, and report the northernmost established population of I. scapularis in Ontario. Corkscrew Island is hyperendemic for Lyme disease and has the highest prevalence of B. burgdorferi s.l. for any established population in Canada. Because of this very high infection prevalence, this population of I. scapularis has likely been established for decades. Of epidemiological significance, cottage owners, island visitors, outdoors enthusiasts, and medical professionals must be vigilant that B. burgdorferi s.l.-infected I. scapularis on Corkscrew Island pose a serious public health risk.
Collapse
Affiliation(s)
- John D Scott
- Lyme Ontario, Research Division, 365 St. David St. South, Fergus, Ontario, Canada N1M 2L7
| | - Janet E Foley
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California 95616, United States of America
| | - Kerry L Clark
- Epidemiology & Environmental Health, Department of Public Health, University of North Florida, 1 UNF Drive, Jacksonville, Florida 32224, United States of America
| | - John F Anderson
- Department of Entomology and Center for Vector Ecology and Zoonotic Diseases. The Connecticut Agricultural Experiment Station, P.O. Box 1106, New Haven, Connecticut 06504-1106, United States of America
| | - Lance A Durden
- Department of Biology, Georgia Southern University, 4324 Old Register Road, Statesboro, Georgia 30458, United States of America
| | - Jodi M Manord
- Epidemiology & Environmental Health, Department of Public Health, University of North Florida, 1 UNF Drive, Jacksonville, Florida 32224, United States of America
| | - Morgan L Smith
- Epidemiology & Environmental Health, Department of Public Health, University of North Florida, 1 UNF Drive, Jacksonville, Florida 32224, United States of America
| |
Collapse
|