1
|
Diving into the complexity of the spirochetal endoflagellum. Trends Microbiol 2023; 31:294-307. [PMID: 36244923 DOI: 10.1016/j.tim.2022.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022]
Abstract
Spirochaetes, a phylum that includes medically important pathogens such as the causative agents of Lyme disease, syphilis, and leptospirosis, are in many ways highly unique bacteria. Their cell morphology, subcellular organization, and metabolism reveal atypical features. Spirochetal motility is also singular, dependent on the presence of periplasmic flagella or endoflagella, inserted subterminally at cell poles and not penetrating the outer membrane and elongating outside the cell as in enterobacteria. In this review we present a comprehensive comparative genomics analysis of endoflagellar systems in spirochetes, highlighting recent findings on the flagellar basal body and filament. Continued progress in understanding the function and architecture of spirochetal flagella is uncovering paradigm-shifting mechanisms of bacterial motility.
Collapse
|
2
|
Orjuela AG, Parra-Arango JL, Sarmiento-Rubiano LA. Bovine leptospirosis: effects on reproduction and an approach to research in Colombia. Trop Anim Health Prod 2022; 54:251. [PMID: 35943610 PMCID: PMC9360731 DOI: 10.1007/s11250-022-03235-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/13/2022] [Indexed: 12/01/2022]
Abstract
Leptospirosis is the most widespread zoonosis worldwide, causing severe effects on beef and dairy cattle farming and other livestock. Colombia geographical location in the tropical zone, high biodiversity, and climatic conditions promote Leptospira growth and prevalence. This review article presents state-of-the-art knowledge about the effects of leptospirosis on bovine reproduction and a critical analysis of the research carried out in Colombia. The analysis of the information allows us to infer a sustained increase in prevalence over the last decade in the densest livestock production areas and a high serovar diversity of circulating pathogenic Leptospira. Given the zoonotic nature of leptospirosis, an inter-institutional effort is required to implement prevention, control, and monitoring programs under one-health concept.
Collapse
Affiliation(s)
- Agustín Góngora Orjuela
- Grupo de Investigación en Reproducción y Genética Animal (Reproduction and Animal Genetics Research Group, GIRGA), Universidad de los Llanos, Villavicencio, Meta Colombia
| | | | | |
Collapse
|
3
|
Campos CL, Gomes LR, Covarrubias AE, Kato EE, Souza GG, Vasconcellos SA, Heinemann MB, Martins EAL, Ho PL, Da Costa RMA, Da Silva JB. A Three-Dimensional Lung Cell Model to Leptospira Virulence Investigations. Curr Microbiol 2022; 79:57. [PMID: 34982247 DOI: 10.1007/s00284-021-02720-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/08/2021] [Indexed: 11/30/2022]
Abstract
Leptospirosis is a worldwide zoonosis and a serious public health threat in tropical and subtropical areas. The etiologic agents of leptospirosis are pathogenic spirochetes from the genus Leptospira. In severe cases, patients develop a pulmonary hemorrhage that is associated with high fatality rates. Several animal models were established for leptospirosis studies, such as rodents, dogs, and monkeys. Although useful to study the relationship among Leptospira and its hosts, the animal models still exhibit economic and ethical limitation reasons and do not fully represent the human infection. As an attempt to bridge the gap between animal studies and clinical information from patients, we established a three-dimensional (3-D) human lung cell culture for Leptospira infection. We show that Leptospira is able to efficiently infect the cell lung spheroids and also to infiltrate in deeper areas of the cell aggregates. The ability to infect the 3-D lung cell aggregates was time-dependent. The 3-D spheroids infection occurred up to 120 h in studies with two serovars, Canicola and Copenhageni. We standardized the number of bacteria in the initial inoculum for infection of the spheroids and we also propose two alternative culture media conditions. This new approach was validated by assessing the expression of three genes of Leptospira related to virulence and motility. The transcripts of these genes increased in both culture conditions, however, in higher rates and earlier times in the 3-D culture. We also assessed the production of chemokines by the 3-D spheroids before and after Leptospira infection, confirming induction of two of them, mainly in the 3-D spheroids. Chemokine CCL2 was expressed only in the 3-D cell culture. Increasing of this chemokine was observed previously in infected animal models. This new approach provides an opportunity to study the interaction of Leptospira with the human lung epithelium in vitro.
Collapse
Affiliation(s)
- Camila L Campos
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Luciana R Gomes
- Laboratório de Ciclo Celular-Center for Research on Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Ambart E Covarrubias
- Facultad de Ciencias de la Salud, Escuela de Tecnología Médica, Universidad San Sebastian, Concepción, Chile
| | - Ellen E Kato
- Laboratorio de Fisiopatologia, Instituto Butantan, São Paulo, Brazil
| | - Gisele G Souza
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Silvio A Vasconcellos
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Marcos B Heinemann
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paulo L Ho
- Divisão BioIndustrial, Instituto Butantan, São Paulo, Brazil
| | - Renata M A Da Costa
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.,Global Antibiotics Research and Development Partnership (GARDP), Chemin Louis-Dunant 15, 1202, Geneva, Switzerland
| | - Josefa B Da Silva
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.
| |
Collapse
|
4
|
Philip N, Jani J, Azhari NN, Sekawi Z, Neela VK. In vivo and in silico Virulence Analysis of Leptospira Species Isolated From Environments and Rodents in Leptospirosis Outbreak Areas in Malaysia. Front Microbiol 2021; 12:753328. [PMID: 34803975 PMCID: PMC8602918 DOI: 10.3389/fmicb.2021.753328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022] Open
Abstract
The zoonotic disease leptospirosis is caused by pathogenic species of the genus Leptospira. With the advancement of studies in leptospirosis, several new species are being reported. It has always been a query, whether Leptospira species, serovars, and strains isolated from different geographical locations contribute to the difference in the disease presentations and severity. In an epidemiological surveillance study performed in Malaysia, we isolated seven novel intermediate and saprophytic species (Leptospira semungkisensis, Leptospira fletcheri, Leptospira langatensis, Leptospira selangorensis, Leptospira jelokensis, Leptospira perdikensis, Leptospira congkakensis) from environments and three pathogenic species from rodents (Leptospira borgpetersenii strain HP364, Leptospira weilii strain SC295, Leptospira interrogans strain HP358) trapped in human leptospirosis outbreak premises. To evaluate the pathogenic potential of these isolates, we performed an in vivo and in silico virulence analysis. Environmental isolates and strain HP364 did not induce any clinical manifestations in hamsters. Strain SC295 caused inactivity and weight loss with histopathological changes in kidneys, however, all hamsters survived until the end of the experiment. Strain HP358 showed a high virulent phenotype as all infected hamsters died or were moribund within 7 days postinfection. Lungs, liver, and kidneys showed pathological changes with hemorrhage as the main presentation. In silico analysis elucidated the genome size of strain HP358 to be larger than strains HP364 and SC295 and containing virulence genes reported in Leptospira species and a high number of specific putative virulence factors. In conclusion, L. interrogans strain HP358 was highly pathogenic with fatal outcome. The constituent of Leptospira genomes may determine the level of disease severity and that needs further investigations.
Collapse
Affiliation(s)
- Noraini Philip
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Jaeyres Jani
- Borneo Medical and Health Research Center, Universiti Malaysia Sabah, Sabah, Malaysia
| | - Nurul Natasya Azhari
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Vasantha Kumari Neela
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
5
|
Correlation between renal distribution of leptospires during the acute phase and chronic renal dysfunction in a hamster model of infection with Leptospira interrogans. PLoS Negl Trop Dis 2021; 15:e0009410. [PMID: 34143778 PMCID: PMC8213162 DOI: 10.1371/journal.pntd.0009410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 04/26/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Leptospirosis has been described as a biphasic disease consisting of hematogenous dissemination to major organs in the acute phase and asymptomatic renal colonization in the chronic phase. Several observational studies have suggested an association between leptospirosis and chronic kidney disease (CKD). We investigated the dynamics of leptospires and histopathological changes in the kidney to understand the relationship between them, and also investigated the extent of renal dysfunction in the acute and chronic phases of leptospirosis using a hamster model. FINDINGS Hamsters (n = 68) were subcutaneously infected with 1 × 104 cells of the Leptospira interrogans serovar Manilae strain UP-MMC-SM. A total of 53 infected hamsters developed fatal acute leptospirosis, and the remaining 15 hamsters recovered from the acute phase, 13 of which showed Leptospira colonization in the kidneys in the chronic phase. Five asymptomatic hamsters also had renal colonization in the chronic phase. Immunofluorescence staining showed that leptospires were locally distributed in the renal interstitium in the early acute phase and then spread continuously into the surrounding interstitium. The kidneys of the surviving hamsters in the chronic phase showed patchy lesions of atrophic tubules, a finding of chronic tubulointerstitial nephritis, which were substantially consistent with the distribution of leptospires in the renal interstitium. The degree of atrophic tubules in kidney sections correlated statistically with the serum creatinine level in the chronic phase (rs = 0.78, p = 0.01). CONCLUSION Subcutaneous infection with pathogenic leptospires could cause acute death or chronic leptospirosis in hamsters after surviving the acute phase. We suggest that the renal distribution of leptospires during the acute phase probably affected the extent of tubular atrophy, leading to CKD.
Collapse
|
6
|
Ge YM, Sun AH, Ojcius DM, Li SJ, Hu WL, Lin X, Yan J. M16-Type Metallopeptidases Are Involved in Virulence for Invasiveness and Diffusion of Leptospira interrogans and Transmission of Leptospirosis. J Infect Dis 2021; 222:1008-1020. [PMID: 32274497 DOI: 10.1093/infdis/jiaa176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Leptospirosis is a global zoonotic infectious disease caused by Leptospira interrogans. The pathogen rapidly invades into hosts and diffuses from bloodstream into internal organs and excretes from urine to cause transmission of leptospirosis. However, the mechanism of leptospiral invasiveness remains poorly understood. METHODS Proteolytic activity of M16-type metallopeptidases (Lep-MP1/2/3) of L. interrogans was determined by spectrophotometry. Expression and secretion of Lep-MP1/2/3 during infection of cells were detected by quantitative reverse-transcription polymerase chain reaction, Western blot assay, and confocal microscopy. Deletion and complementation mutants of the genes encoding Lep-MP1/2/3 were generated to determine the roles of Lep-MP1/2/3 in invasiveness using transwell assay and virulence in hamsters. RESULTS Leptospira interrogans but not saprophytic Leptospira biflexa strains were detectable for Lep-MP-1/2/3-encoding genes. rLep-MP1/2/3 hydrolyzed extracellular matrix proteins, but rLep-MP1/3 displayed stronger proteolysis than rLep-MP2, with 123.179/340.136 μmol/L Km and 0.154/0.159 s-1 Kcat values. Expression, secretion and translocation of Lep-MP1/2/3 during infection of cells were increased. ΔMP1/3 but not ΔMP2 mutant presented attenuated transmigration through cell monolayers, decreased leptospiral loading in the blood, lungs, liver, kidneys, and urine, and 10/13-fold decreased 50% lethal dose and milder histopathologic injury in hamsters. CONCLUSIONS Lep-MP1 and 3 are involved in virulence of L. interrogans in invasion into hosts and diffusion in vivo, and transmission of leptospirosis.
Collapse
Affiliation(s)
- Yu-Mei Ge
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Ai-Hua Sun
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - David M Ojcius
- Department of Biomedical Sciences, School of Dentistry, University of the Pacific, San Francisco, California, USA.,Université de Paris, Paris, France
| | - Shi-Jun Li
- Guizhou Provincial Centre for Disease Control and Prevention, Guiyang, Guizhou, People's Republic of China
| | - Wei-Lin Hu
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Xu'ai Lin
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
7
|
Kaur H, Kalia M, Singh V, Modgil V, Mohan B, Taneja N. In silico identification and characterization of promising drug targets in highly virulent uropathogenic Escherichia coli strain CFT073 by protein-protein interaction network analysis. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
8
|
Whole genome sequencing and de novo assembly of three virulent Indian isolates of Leptospira. INFECTION GENETICS AND EVOLUTION 2020; 85:104579. [PMID: 33017688 DOI: 10.1016/j.meegid.2020.104579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/09/2020] [Accepted: 09/30/2020] [Indexed: 12/26/2022]
Abstract
Leptospirosis is a re-emerging bacterial zoonosis caused by pathogenic Leptospira, with a worldwide distribution and becoming a major public health concern. Prophylaxis of this disease is difficult due to several factors such as non-specific variable clinical manifestation, presence of a large number of serovar, species and asymptomatic reservoir hosts, lack of proper diagnostics and vaccines. Despite its global importance and severity of the disease, knowledge about the molecular mechanism of pathogenesis and evolution of pathogenic species of Leptospira remains limited. In this study, we sequenced and analyzed three highly pathogenic species of Indian isolates of Leptospira (interrogans, santarosai, and kirschneri). Additionally, we identified some virulence-related and CRISPR-Cas genes. The virulent analysis showed 232 potential virulence factors encoding proteins in L. interrogans strain Salinem and L. santarosai strain M-4 genome. While the genome of L. kirschneri strain Wumalasena was predicted to encode 198 virulence factor proteins. The variant calling analysis revealed 1151, 19,786, and 22,996 single nucleotide polymorphisms (SNPs) for L. interrogans strain Salinem, L. kirschneri strain Wumalasena and L. santarosai strain M-4, respectively, with a maximum of 5315 missense and 12,221 synonymous mutations for L. santarosai strain M-4. The structural analyses of genomes indicated potential evidence of inversions and structural rearrangment in all three genomes. The availability of these genome sequences and in silico analysis of Leptospira will provide a basis for a deeper understanding of their molecular diversity and pathogenesis mechanism, and further pave a way towards proper management of the disease.
Collapse
|
9
|
Kędzierska-Mieszkowska S, Arent Z. AAA+ Molecular Chaperone ClpB in Leptospira interrogans: Its Role and Significance in Leptospiral Virulence and Pathogenesis of Leptospirosis. Int J Mol Sci 2020; 21:E6645. [PMID: 32932775 PMCID: PMC7555560 DOI: 10.3390/ijms21186645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Bacterial ClpB is an ATP-dependent disaggregase that belongs to the Hsp100/Clp subfamily of the AAA+ ATPases and cooperates with the DnaK chaperone system in the reactivation of aggregated proteins, as well as promotes bacterial survival under adverse environmental conditions, including thermal and oxidative stresses. In addition, extensive evidence indicates that ClpB supports the virulence of numerous bacteria, including pathogenic spirochaete Leptospira interrogans responsible for leptospirosis in animals and humans. However, the specific function of ClpB in leptospiral virulence still remains to be fully elucidated. Interestingly, ClpB was predicted as one of the L. interrogans hub proteins interacting with human proteins, and pathogen-host protein interactions are fundamental for successful invasion of the host immune system by bacteria. The aim of this review is to discuss the most important aspects of ClpB's function in L. interrogans, including contribution of ClpB to leptospiral virulence and pathogenesis of leptospirosis, a zoonotic disease with a significant impact on public health worldwide.
Collapse
Affiliation(s)
| | - Zbigniew Arent
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, 30-059 Krakow, Poland;
| |
Collapse
|
10
|
Murillo A, Goris M, Ahmed A, Cuenca R, Pastor J. Leptospirosis in cats: Current literature review to guide diagnosis and management. J Feline Med Surg 2020; 22:216-228. [PMID: 32093581 PMCID: PMC11132596 DOI: 10.1177/1098612x20903601] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
GLOBAL IMPORTANCE Leptospirosis is the most widespread zoonosis worldwide. Mammals (eg, rats, horses, cows, pigs, dogs, cats and aquatic species, such as sea lions and northern elephant seals) can all be infected by leptospires. Infection in animals occurs through contact with urine or water contaminated with the bacteria. In people, the disease is acquired mainly from animal sources or through recreational activities in contaminated water. PRACTICAL RELEVANCE Literature on the clinical presentation of leptospirosis in cats is scarce, although it has been demonstrated that cats are susceptible to infection and are capable of developing antibodies. The prevalence of antileptospiral antibodies in cats varies from 4% to 33.3% depending on the geographical location. Urinary shedding of leptospires in naturally infected cats has been reported, with a prevalence of up to 68%. Infection in cats has been associated with the consumption of infected prey, especially rodents. Thus, outdoor cats have a higher risk of becoming infected. CLINICAL CHALLENGES Clinical presentation of this disease in cats is rare and it is not known what role cats have in the transmission of leptospirosis. Ongoing work is needed to characterise feline leptospirosis. AUDIENCE This review is aimed at all veterinarians, both general practitioners who deal with cats on a daily basis in private practice, as well as feline practitioners, since both groups face the challenge of diagnosing and treating infectious and zoonotic diseases. EVIDENCE BASE The current literature on leptospirosis in cats is reviewed. To date, few case reports have been published in the field, and information has mostly been extrapolated from infections in people and dogs. This review is expected to serve as a guide for the diagnosis and management of the disease in cats.
Collapse
Affiliation(s)
- Andrea Murillo
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Marga Goris
- OIE and National Collaborating Centre for Reference and Research on Leptospirosis (NRL), Amsterdam UMC, University of Amsterdam, Medical Microbiology, Meibergdreef 39, 1105 AZ, Amsterdam, Netherlands
| | - Ahmed Ahmed
- OIE and National Collaborating Centre for Reference and Research on Leptospirosis (NRL), Amsterdam UMC, University of Amsterdam, Medical Microbiology, Meibergdreef 39, 1105 AZ, Amsterdam, Netherlands
| | - Rafaela Cuenca
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Josep Pastor
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
11
|
Sun AH, Liu XX, Yan J. Leptospirosis is an invasive infectious and systemic inflammatory disease. Biomed J 2020; 43:24-31. [PMID: 32200953 PMCID: PMC7090314 DOI: 10.1016/j.bj.2019.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/15/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023] Open
Abstract
Pathogenic Leptospira species are the causative agents of leptospirosis, a world-spreading zoonotic infectious disease. The pathogens possess a powerful invasiveness by invading human body through mucosal/skin barriers, rapid entry into bloodstream to cause septicemia, diffusion from bloodstream into internal organs and tissues to cause aggravation of disease, and discharge from urine through renal tubules to form natural infectious sources. Leptospirosis patients present severe inflammatory symptoms such as high fever, myalgia and lymphadenectasis. Hemorrhage and jaundice are the pathological features of this disease. Previous studies revealed that some outer membrane proteins of Leptospira interrogans, the most important pathogenic Leptospira species, acted as adherence factors to binding to receptor molecules (fibronectin, laminin and collagens) in extracellular matrix of host cells. Collagenase, metallopeptidases and endoflagellum contributed to the invasiveness of L. interrogans. Except for lipopolysaccharide, multiple hemolysins of L. interrogans displayed a powerful ability to induce pro-inflammatory cytokines and hepatocyte apoptosis. vWA and platelet activating factor acetylhydrolase-like proteins from L. interrogans could induce severe pulmonary hemorrhage in mice. L. interrogans utilized cellular endocytic recycling and vesicular transport systems for intracellular migration and transcellular transport. All the research achievements are helpful for further understanding the virulence of pathogenic Leptospira species and pathogenesis of leptospirosis.
Collapse
Affiliation(s)
- Ai-Hua Sun
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, PR China
| | - Xiao-Xiang Liu
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, PR China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
12
|
Henderson LD, Matthews-Palmer TRS, Gulbronson CJ, Ribardo DA, Beeby M, Hendrixson DR. Diversification of Campylobacter jejuni Flagellar C-Ring Composition Impacts Its Structure and Function in Motility, Flagellar Assembly, and Cellular Processes. mBio 2020; 11:e02286-19. [PMID: 31911488 PMCID: PMC6946799 DOI: 10.1128/mbio.02286-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022] Open
Abstract
Bacterial flagella are reversible rotary motors that rotate external filaments for bacterial propulsion. Some flagellar motors have diversified by recruiting additional components that influence torque and rotation, but little is known about the possible diversification and evolution of core motor components. The mechanistic core of flagella is the cytoplasmic C ring, which functions as a rotor, directional switch, and assembly platform for the flagellar type III secretion system (fT3SS) ATPase. The C ring is composed of a ring of FliG proteins and a helical ring of surface presentation of antigen (SPOA) domains from the switch proteins FliM and one of two usually mutually exclusive paralogs, FliN or FliY. We investigated the composition, architecture, and function of the C ring of Campylobacter jejuni, which encodes FliG, FliM, and both FliY and FliN by a variety of interrogative approaches. We discovered a diversified C. jejuni C ring containing FliG, FliM, and both FliY, which functions as a classical FliN-like protein for flagellar assembly, and FliN, which has neofunctionalized into a structural role. Specific protein interactions drive the formation of a more complex heterooligomeric C. jejuni C-ring structure. We discovered that this complex C ring has additional cellular functions in polarly localizing FlhG for numerical regulation of flagellar biogenesis and spatial regulation of division. Furthermore, mutation of the C. jejuni C ring revealed a T3SS that was less dependent on its ATPase complex for assembly than were other systems. Our results highlight considerable evolved flagellar diversity that impacts motor output, biogenesis, and cellular processes in different species.IMPORTANCE The conserved core of bacterial flagellar motors reflects a shared evolutionary history that preserves the mechanisms essential for flagellar assembly, rotation, and directional switching. In this work, we describe an expanded and diversified set of core components in the Campylobacter jejuni flagellar C ring, the mechanistic core of the motor. Our work provides insight into how usually conserved core components may have diversified by gene duplication, enabling a division of labor of the ancestral protein between the two new proteins, acquisition of new roles in flagellar assembly and motility, and expansion of the function of the flagellum beyond motility, including spatial regulation of cell division and numerical control of flagellar biogenesis in C. jejuni Our results highlight that relatively small changes, such as gene duplications, can have substantial ramifications on the cellular roles of a molecular machine.
Collapse
Affiliation(s)
- Louie D Henderson
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Connor J Gulbronson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Deborah A Ribardo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Morgan Beeby
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
13
|
Gupta R, Verma R, Pradhan D, Jain AK, Umamaheswari A, Rai CS. An in silico approach towards identification of novel drug targets in pathogenic species of Leptospira. PLoS One 2019; 14:e0221446. [PMID: 31430340 PMCID: PMC6701809 DOI: 10.1371/journal.pone.0221446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/06/2019] [Indexed: 11/18/2022] Open
Abstract
Leptospirosis is one of the leading zoonotic infections worldwide. As with other infectious diseases, report of antimicrobial resistance to existing therapeutic arsenal poses challenges in the management of disease. Hence, identification of novel drug targets for the pathogen deems essential. Present study used combined approach of comparative and subtractive genomics to identify putative drug targets. Crucial genes of 16 pathogenic Leptospira strains were filtered and subjected to homology search via target identification tool "TiD". Thereafter, comparative analysis was performed for non-homologous, essential genes to accomplish the broad-spectrum drug target. Consequently, 37 essential genes were found to be conserved in at least 10 strains of Leptospira. Further, prioritization of resultant set of genes revealed 18 were hubs in protein-protein interaction network. Sixteen putative targets among the hub genes were conserved in all strains of Leptospira. Out of sixteen, fourteen were enzymes while 8 were novel and 4 were involved in virulence mechanism. In addition, genome scale metabolic network reconstruction and choke point analysis revealed cobA (porphyrin and chlorophyll metabolism) and thiL (thiamine metabolism) as chokepoints in their respective metabolic pathways. The proposed hub genes could act as putative broad-spectrum drug targets for Leptospira species, however, these putative targets should be validated to ensure them as real one prior to utilizing them for target based lead discovery.
Collapse
Affiliation(s)
- Reena Gupta
- University School of Information, Communication & Technology, Guru Gobind Singh Indraprastha University, New Delhi, India
| | - Rashi Verma
- Biomedical Informatics Centre, National Institute of Pathology-Indian Council of Medical Research, New Delhi, India
| | - Dibyabhaba Pradhan
- Computational Genomics Centre, Indian Council of Medical Research, Campus—All India Institute of Medical Sciences, New Delhi, India
| | - Arun Kumar Jain
- Biomedical Informatics Centre, National Institute of Pathology-Indian Council of Medical Research, New Delhi, India
| | - Amineni Umamaheswari
- Department of Bioinformatics, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Chandra Shekhar Rai
- University School of Information, Communication & Technology, Guru Gobind Singh Indraprastha University, New Delhi, India
| |
Collapse
|
14
|
Abstract
Until about 15 years ago, the molecular and cellular basis for pathogenesis in leptospirosis was virtually unknown. The determination of the first full genome sequence in 2003 was followed rapidly by other whole genome sequences, whose availability facilitated the development of transposon mutagenesis and then directed mutagenesis of pathogenic Leptospira spp. The combination of genomics, transcriptomics and mutant construction and characterisation has resulted in major progress in our understanding of the taxonomy and biology of Leptospira. The most recent advances are analysed and discussed in this chapter.
Collapse
Affiliation(s)
- Dieter Bulach
- Melbourne Bioinformatics, The University of Melbourne, Carlton, VIC, Australia
| | - Ben Adler
- Department of Microbiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
15
|
Abstract
This chapter covers the progress made in the Leptospira field since the application of mutagenesis techniques and how they have allowed the study of virulence factors and, more generally, the biology of Leptospira. The last decade has seen advances in our ability to perform molecular genetic analysis of Leptospira. Major achievements include the generation of large collections of mutant strains and the construction of replicative plasmids, enabling complementation of mutations. However, there are still no practical tools for routine genetic manipulation of pathogenic Leptospira strains, slowing down advances in pathogenesis research. This review summarizes the status of the molecular genetic toolbox for Leptospira species and highlights new challenges in the nascent field of Leptospira genetics.
Collapse
Affiliation(s)
- Mathieu Picardeau
- Biology of Spirochetes Unit, Institut Pasteur, 28 Rue Du Docteur Roux, 75724, Paris Cedex 15, France.
| |
Collapse
|
16
|
Li Y, Li KX, Hu WL, Ojcius DM, Fang JQ, Li SJ, Lin X, Yan J. Endocytic recycling and vesicular transport systems mediate transcytosis of Leptospira interrogans across cell monolayer. eLife 2019; 8:44594. [PMID: 31012847 PMCID: PMC6513555 DOI: 10.7554/elife.44594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/18/2019] [Indexed: 12/18/2022] Open
Abstract
Many bacterial pathogens can cause septicemia and spread from the bloodstream into internal organs. During leptospirosis, individuals are infected by contact with Leptospira-containing animal urine-contaminated water. The spirochetes invade internal organs after septicemia to cause disease aggravation, but the mechanism of leptospiral excretion and spreading remains unknown. Here, we demonstrated that Leptospira interrogans entered human/mouse endothelial and epithelial cells and fibroblasts by caveolae/integrin-β1-PI3K/FAK-mediated microfilament-dependent endocytosis to form Leptospira (Lep)-vesicles that did not fuse with lysosomes. Lep-vesicles recruited Rab5/Rab11 and Sec/Exo-SNARE proteins in endocytic recycling and vesicular transport systems for intracellular transport and release by SNARE-complex/FAK-mediated microfilament/microtubule-dependent exocytosis. Both intracellular leptospires and infected cells maintained their viability. Leptospiral propagation was only observed in mouse fibroblasts. Our study revealed that L. interrogans utilizes endocytic recycling and vesicular transport systems for transcytosis across endothelial or epithelial barrier in blood vessels or renal tubules, which contributes to spreading in vivo and transmission of leptospirosis.
Collapse
Affiliation(s)
- Yang Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai-Xuan Li
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Lin Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - David M Ojcius
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, United States
| | - Jia-Qi Fang
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shi-Jun Li
- Institute of Communicable Disease Prevention and Control, Guizhou Provincial Centre for Disease Control and Prevention, Guiyang, China
| | - Xu'ai Lin
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yan
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Kurilung A, Keeratipusana C, Suriyaphol P, Hampson DJ, Prapasarakul N. Genomic analysis of Leptospira interrogans serovar Paidjan and Dadas isolates from carrier dogs and comparative genomic analysis to detect genes under positive selection. BMC Genomics 2019; 20:168. [PMID: 30832578 PMCID: PMC6399948 DOI: 10.1186/s12864-019-5562-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/25/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Leptospirosis is an emerging infectious disease worldwide that can cause high morbidity and mortality rates in humans and animals. The causative spirochetes have reservoirs in mammalian hosts, but there has been limited analysis of the genomes of isolates recovered from animals. The aims of this study were to characterize genomic features of two Leptospira interrogans strains recently isolated from asymptomatic dogs in Thailand (strains CUDO5 and CDUO8), and to perform comparative genome analyses with other strains. Molecular adaptive evolution in L. interrogans as signaled by positive selection also was analyzed. RESULTS Whole genome sequence analysis revealed that strains CUDO5 and CUDO8 had genome sizes of approximately 4.9 Mbp with 35.1% GC contents. Using monoclonal antibodies, strains CUDO5 and CUDO8 were identified as serovars Paidjan and Dadas, respectively. These strains harbored genes known to be associated with acute and chronic infections. Using Single Nucleotide Polymorphisms phylogeny (SNPs) with 97 L. interrogans strains, CUDO5 and CUDO8 had closest genetic relatedness with each other. Nevertheless, the serovar determinant region (rfb locus) showed variations in the genes encoding sugar biosynthesis. Amongst 13 representative L. interrogans strains examined for molecular adaptive evolution through positive selection under the site-model of Phylogenetic Analysis of Maximum Likelihood, genes responsible for iron acquisition (tlyA and hbpA), motility (fliN2, flgK, and flhB) and thermal adaptation (lpxD1) were under increased selective pressure. CONCLUSIONS L. interrogans serovar Paidjan strain CUDO5 and serovar Dadas strain CUDO8 had close genetic relatedness as analyzed by SNPs phylogeny. They contained genes with established roles in acute and chronic leptospirosis. The rfb locus in both serovars showed gene variation associated with sugar biosynthesis. Positive selection analysis indicated that genes encoding factors involved in motility, temperature adaptation, and iron acquisition were under strong positive selection in L. interrogans. These may be associated with adaptation in the early stages of infection.
Collapse
Affiliation(s)
- Alongkorn Kurilung
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Chantisa Keeratipusana
- Bioinformatics and Data Management for Research Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Prapat Suriyaphol
- Bioinformatics and Data Management for Research Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - David J. Hampson
- Department of Infectious Diseases and Public Health, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Nuvee Prapasarakul
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Diagnosis and Monitoring of Animal Pathogens Research Unit, Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
18
|
A novel Fas-binding outer membrane protein and lipopolysaccharide of Leptospira interrogans induce macrophage apoptosis through the Fas/FasL-caspase-8/-3 pathway. Emerg Microbes Infect 2018; 7:135. [PMID: 30061622 PMCID: PMC6066479 DOI: 10.1038/s41426-018-0135-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 06/23/2018] [Indexed: 02/08/2023]
Abstract
Leptospira interrogans is the major causative agent of leptospirosis, an emerging, globally spreading zoonotic infectious disease. The pathogen induces macrophage apoptosis, but the molecular basis and mechanism remain unknown. In the present study, we found that L. interrogans caused apoptosis of phagocytosis-inhibited macrophages, and the product of the L. interrogans LB047 gene (Lep-OMP047) was the unique protein captured by mouse and human Fas proteins. The recombinant expressed Lep-OMP047 (rLep-OMP047) strongly bound mouse and human Fas proteins with equilibrium association constant (KD) values of 5.20 × 10−6 to 2.84 × 10−9 M according to surface plasmon resonance measurement and isothermal titration calorimetry. Flow-cytometric examination showed that 5 μg rLep-OMP047 or 1 μg lipopolysaccharide of L. interrogans (Lep-LPS) caused 43.70% or 21.90% early apoptosis in mouse J774A.1 macrophages and 28.41% or 15.80% for PMA-differentiated human THP-1 macrophages, respectively, but the apoptosis was blocked by Fas-antagonizing IgGs, Fas siRNAs, and caspase-8/-3 inhibitors. Moreover, Lep-OMP047 was significantly upregulated during infection of macrophages. Lep-LPS promoted the expression and cytomembrane translocation of Fas and FasL in macrophages. The JNK and p38 MAPK but not ERK signaling pathways, as well as the transcription factors c-Jun and ATF2 but not CHOP, mediated Lep-LPS-induced Fas/FasL expression and translocation. TLR2 but not TLR4 mediated Lep-LPS-induced JNK/p38 MAPK activation. Therefore, we demonstrated that a novel Fas-binding OMP and LPS of L. interrogans induce macrophage apoptosis through the Fas/FasL-caspase-8/-3 pathway.
Collapse
|
19
|
Wunder EA, Slamti L, Suwondo DN, Gibson KH, Shang Z, Sindelar CV, Trajtenberg F, Buschiazzo A, Ko AI, Picardeau M. FcpB Is a Surface Filament Protein of the Endoflagellum Required for the Motility of the Spirochete Leptospira. Front Cell Infect Microbiol 2018; 8:130. [PMID: 29868490 PMCID: PMC5953323 DOI: 10.3389/fcimb.2018.00130] [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: 01/26/2018] [Accepted: 04/13/2018] [Indexed: 12/30/2022] Open
Abstract
The spirochete endoflagellum is a unique motility apparatus among bacteria. Despite its critical importance for pathogenesis, the full composition of the flagellum remains to be determined. We have recently reported that FcpA is a novel flagellar protein and a major component of the sheath of the filament of the spirochete Leptospira. By screening a library of random transposon mutants in the spirochete Leptospira biflexa, we found a motility-deficient mutant harboring a disruption in a hypothetical gene of unknown function. Here, we show that this gene encodes a surface component of the endoflagellar filament and is required for typical hook- and spiral-shaped ends of the cell body, coiled structure of the endoflagella, and high velocity phenotype. We therefore named the gene fcpB for flagellar-coiling protein B. fcpB is conserved in all members of the Leptospira genus, but not present in other organisms including other spirochetes. Complementation of the fcpB- mutant restored the wild-type morphology and motility phenotypes. Immunoblotting with anti-FcpA and anti-FcpB antisera and cryo-electron microscopy of the filament indicated that FcpB assembled onto the surface of the sheath of the filament and mostly located on the outer (convex) side of the coiled filament. We provide evidence that FcpB, together with FcpA, are Leptospira-specific novel components of the sheath of the filament, key determinants of the coiled and asymmetric structure of the endoflagella and are essential for high velocity. Defining the components of the endoflagella and their functions in these atypical bacteria should greatly enhance our understanding of the mechanisms by which these bacteria produce motility.
Collapse
Affiliation(s)
- Elsio A Wunder
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States.,Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Brazil
| | - Leyla Slamti
- Institut Pasteur, Biology of Spirochetes Unit, Paris, France
| | - David N Suwondo
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Kimberley H Gibson
- Departments of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States
| | - Zhiguo Shang
- Departments of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States
| | - Charles V Sindelar
- Departments of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States
| | - Felipe Trajtenberg
- Institut Pasteur de Montevideo, Laboratory of Molecular and Structural Microbiology, Montevideo, Uruguay
| | - Alejandro Buschiazzo
- Institut Pasteur de Montevideo, Laboratory of Molecular and Structural Microbiology, Montevideo, Uruguay.,Department of Microbiology, Institut Pasteur, Paris, France
| | - Albert I Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States.,Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Brazil
| | | |
Collapse
|
20
|
Isolation and Identification of Putative Protein Substrates of the AAA+ Molecular Chaperone ClpB from the Pathogenic Spirochaete Leptospira interrogans. Int J Mol Sci 2018; 19:ijms19041234. [PMID: 29670056 PMCID: PMC5979558 DOI: 10.3390/ijms19041234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/13/2018] [Accepted: 04/15/2018] [Indexed: 01/22/2023] Open
Abstract
Bacterial ClpB is an ATP-dependent Hsp100 chaperone that reactivates aggregated proteins in cooperation with the DnaK chaperone system and promotes survival of bacteria under stress conditions. A large number of publications also indicate that ClpB supports the virulence of bacteria, including a pathogenic spirochaete Leptospira interrogans responsible for leptospirosis in both animals and humans. However, the exact role of ClpB in bacterial pathogenicity remains poorly characterized. It can be assumed that ClpB, due to its role as the molecular chaperone, mediates refolding of essential bacterial proteins, including the known virulence factors, which may become prone to aggregation under infection-induced stresses. In this study, we identified putative substrates of ClpB from L. interrogans (ClpBLi). For this purpose, we used a proteomic approach combining the ClpB-Trap affinity pull-down assays, Liquid chromatography-tandem mass spectrometry (LC-MS-MS/MS), and bioinformatics analyses. Most of the identified proteins were enzymes predominantly associated with major metabolic pathways like the tricarboxylic acid (TCA) cycle, glycolysis–gluconeogenesis and amino acid and fatty acid metabolism. Based on our proteomic study, we suggest that ClpB can support the virulence of L.interrogans by protecting the conformational integrity and catalytic activity of multiple metabolic enzymes, thus maintaining energy homeostasis in pathogen cells.
Collapse
|
21
|
Cheng C, Wang H, Ma T, Han X, Yang Y, Sun J, Chen Z, Yu H, Hang Y, Liu F, Fang W, Jiang L, Cai C, Song H. Flagellar Basal Body Structural Proteins FlhB, FliM, and FliY Are Required for Flagellar-Associated Protein Expression in Listeria monocytogenes. Front Microbiol 2018; 9:208. [PMID: 29487588 PMCID: PMC5816908 DOI: 10.3389/fmicb.2018.00208] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/30/2018] [Indexed: 01/08/2023] Open
Abstract
Listeria monocytogenes is a food-associated bacterium that is responsible for food-related illnesses worldwide. In the L. monocytogenes EGD-e genome, FlhB, FliM, and FliY (encoded by lmo0679, lmo0699, and lmo0700, respectively) are annotated as putative flagella biosynthesis factors, but their functions remain unknown. To explore whether FlhB, FliM, and FliY are involved in Listeria flagella synthesis, we constructed flhB, fliM, fliY, and other flagellar-related gene deletion mutants using a homologous recombination strategy. Then, we analyzed the motility, flagella synthesis, and protein expression of these mutant strains. Motility and flagella synthesis were completely abolished in the absence of flhB, fliM, or fliY. These impaired phenotypes were fully restored in the complemented strains CΔflhB, CΔfliM, and CΔfliY. The transcriptional levels of flagellar-related genes, including flaA, fliM, fliY, lmo0695, lmo0698, fliI, and fliS, were downregulated markedly in the absence of flhB, fliM, or fliY. Deletion of flhB resulted in the complete abolishment of FlaA expression, while it decreased FliM and FliY expression. The expression of FlaA was abolished completely in the absence of fliM or fliY. No significant changes were found in the expression of FlhF and two flagella synthesis regulatory factors, MogR and GmaR. We demonstrate for the first time that FlhB, FliM, and FliY not only mediate Listeria motility, but also are involved in regulating flagella synthesis. This study provides novel insights that increase our understanding of the roles played by FlhB, FliM, and FliY in the flagellar type III secretion system in L. monocytogenes.
Collapse
Affiliation(s)
- Changyong Cheng
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China.,Zhejiang University Institute of Preventive Veterinary Medicine and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, China
| | - Hang Wang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Tiantian Ma
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Xiao Han
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Yongchun Yang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Jing Sun
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Zhongwei Chen
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Huifei Yu
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Yi Hang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Fengdan Liu
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| | - Weihuan Fang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China.,Zhejiang University Institute of Preventive Veterinary Medicine and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, China
| | - Lingli Jiang
- Department of Health Services and Management, Ningbo College of Health Sciences, Ningbo, China
| | - Chang Cai
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China.,School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Houhui Song
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Lin'an, China
| |
Collapse
|
22
|
Dong SL, Hu WL, Ge YM, Ojcius DM, Lin X, Yan J. A leptospiral AAA+ chaperone-Ntn peptidase complex, HslUV, contributes to the intracellular survival of Leptospira interrogans in hosts and the transmission of leptospirosis. Emerg Microbes Infect 2017; 6:e105. [PMID: 29184154 PMCID: PMC5717094 DOI: 10.1038/emi.2017.93] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/17/2017] [Accepted: 09/24/2017] [Indexed: 12/13/2022]
Abstract
Leptospirosis caused by Leptospira is a zoonotic disease of global importance but it is considered as an emerging or re-emerging infectious disease in many areas in the world. Until now, the mechanisms about pathogenesis and transmission of Leptospira remains poorly understood. As eukaryotic and prokaryotic proteins can be denatured in adverse environments and chaperone-protease/peptidase complexes degrade these harmful proteins, we speculate that infection may also cause leptospiral protein denaturation, and the HslU and HslV proteins of L. interrogans may compose a complex to degrade denatured proteins that enhances leptospiral survival in hosts. Here we show that leptospiral HslUV is an ATP-dependent chaperone-peptidase complex containing ATPase associated with various cellular activity (AAA+) and N-terminal nucleophile (Ntn) hydrolase superfamily domains, respectively, which hydrolyzed casein and chymotrypsin-like substrates, and this hydrolysis was blocked by threonine protease inhibitors. The infection of J774A.1 macrophages caused the increase of leptospiral denatured protein aggresomes, but more aggresomes accumulated in hslUV gene-deleted mutant. The abundant denatured leptospiral proteins are involved in ribosomal structure, flagellar assembly, two-component signaling systems and transmembrane transport. Compared to the wild-type strain, infection of cells in vitro with the mutant resulted in a higher number of dead leptospires, less leptospiral colony-forming units and lower growth ability, but also displayed a lower half lethal dose, attenuated histopathological injury and decreased leptospiral loading in lungs, liver, kidneys, peripheral blood and urine in hamsters. Therefore, our findings confirmed that HslUV AAA+ chaperone-Ntn peptidase complex of L. interrogans contributes to leptospiral survival in hosts and transmission of leptospirosis.
Collapse
Affiliation(s)
- Shi-Lei Dong
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.,Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Wei-Lin Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yu-Mei Ge
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA 94103, USA
| | - Xu'ai Lin
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jie Yan
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.,Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
23
|
Zaza G, Dalla Gassa A, Felis G, Granata S, Torriani S, Lupo A. Impact of maintenance immunosuppressive therapy on the fecal microbiome of renal transplant recipients: Comparison between an everolimus- and a standard tacrolimus-based regimen. PLoS One 2017; 12:e0178228. [PMID: 28542523 PMCID: PMC5443527 DOI: 10.1371/journal.pone.0178228] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The gut microbiome is the full set of microbes living in the gastrointestinal tract and is emerging as an important dynamic/fluid system that, if altered by environmental, dietetic or pharmacological factors, could considerably influence drug response. However, the immunosuppressive drug-induced modifications of this system are still poorly defined. METHODS We employed an innovative bioinformatics approach to assess differences in the whole-gut microbial metagenomic profile of 20 renal transplant recipients undergoing maintenance treatment with two different immunosuppressive protocols. Nine patients were treated with everolimus plus mycophenolate mofetil (EVE+MMF group), and 11 patients were treated with a standard therapy with tacrolimus plus mycophenolate mofetil (TAC+MMF group). RESULTS A statistical analysis of comparative high-throughput data demonstrated that although similar according to the degree of Shannon diversity (alpha diversity) at the taxonomic level, three functional genes clearly discriminated EVE+MMF versus TAC+MMF (cutoff: log2 fold change≥1, FDR≤0.05). Flagellar motor switch protein (fliNY) and type IV pilus assembly protein pilM (pilM) were significantly enriched in TAC+MMF-treated patients, while macrolide transport system mrsA (msrA) was more abundant in patients treated with EVE+MMF. Finally, PERMANOVA revealed that among the variables analyzed and included in our model, only the consumption of sugar significantly influenced beta diversity. CONCLUSIONS Our study, although performed on a relatively small number of patients, showed, for the first time, specific immunosuppressive-related effects on fecal microbiome of renal transplant recipients and it suggested that the analysis of the gut microbes community could represent a new tool to better understand the effects of drugs currently employed in organ transplantations. However, multicenter studies including healthy controls should be undertaken to better address this objective.
Collapse
Affiliation(s)
- Gianluigi Zaza
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
- * E-mail:
| | | | - Giovanna Felis
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Sandra Torriani
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| |
Collapse
|
24
|
Gomes-Solecki M, Santecchia I, Werts C. Animal Models of Leptospirosis: Of Mice and Hamsters. Front Immunol 2017; 8:58. [PMID: 28270811 PMCID: PMC5318464 DOI: 10.3389/fimmu.2017.00058] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/16/2017] [Indexed: 01/27/2023] Open
Abstract
Pathogenic Leptospira sp. are spirochetal bacteria responsible for leptospirosis, an emerging worldwide zoonosis. These spirochetes are very successful pathogens that infect a wide range of hosts such as fish, reptiles, birds, marsupials, and mammals. Transmission occurs when chronically infected animals excrete live bacteria in their urine, contaminating the environment. Leptospira sp. enter their hosts through damaged skin and mucosa. Chronically infected rats and mice are asymptomatic and are considered as important reservoirs of the disease. Infected humans may develop either a flu-like, usually mild illness with or without chronic asymptotic renal colonization, or a severe acute disease with kidney, liver, and heart failure, potentially leading to death. Leptospirosis is an economic burden on society due to health-care costs related to elevated morbidity of humans and loss of animals of agricultural interest. There are no effective vaccines against leptospirosis. Leptospira sp. are difficult to genetically manipulate which delays the pace of research progress. In this review, we discuss in an historical perspective how animal models have contributed to further our knowledge of leptospirosis. Hamsters, guinea pigs, and gerbils have been instrumental to study the pathophysiology of acute lethal leptospirosis and the Leptospira sp. genes involved in virulence. Chronic renal colonization has been mostly studied using experimentally infected rats. A special emphasis will be placed on mouse models, long thought to be irrelevant since they survive lethal infection. However, mice have recently been shown to be good models of sublethal infection leading to chronic colonization. Furthermore, congenic and transgenic mice have proven essential to study how innate immune cells interact with the pathogen and to understand the role of the toll-like receptor 4, which is important to control Leptospira sp. load and disease. The use of inbred and transgenic mouse models opens up the field to the comprehensive study of immune responses to Leptospira sp. infection and subsequent pathophysiology of inflammation. It also allows for testing of drugs and vaccines in a biological system that can avail of a wealth of molecular tools that enable understanding of the mechanisms of action of protective vaccines.
Collapse
Affiliation(s)
- Maria Gomes-Solecki
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center , Memphis, TN , USA
| | - Ignacio Santecchia
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France; INSERM, équipe Avenir, Paris, France
| | - Catherine Werts
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France; INSERM, équipe Avenir, Paris, France
| |
Collapse
|
25
|
Xu Y, Zheng H, Zhang Y, Wang Y, Zhang J, Li Z, Cui S, Xin X, Ye Q, Chang YF, Wang J. Genomic Analysis of a New Serovar of Leptospira weilii Serogroup Manhao. Front Microbiol 2017; 8:149. [PMID: 28210253 PMCID: PMC5288384 DOI: 10.3389/fmicb.2017.00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/20/2017] [Indexed: 12/29/2022] Open
Abstract
Leptospirosis, caused by pathogenic Leptospira spp., is recognized as an important emerging zoonotic disease throughout the world. In this study, multiple approaches were used to characterize the recently discovered serovar Heyan strain L231. This strain can infect guinea pigs and belonged to the pathogenic species L. weilii. Genome sequencing analysis revealed the draft genome of 4.2 M bp with a G+C content of 40.67% for strain L231, and a total of 4,794 ORFs were identified. The strain L231 genome was found to have a larger LPS biosynthesis locus than that of strains L. interrogans serovar Lai and L. borgpetersenii serovar Hardjobovis. Phylogenomic reconstructions showed that the evolutionary position of L. weilii serovar Heyan was different from that of other serovars from serogroup Manhao. These findings may lead us to a better understanding of Leptospira pathogenesis and evolution.
Collapse
Affiliation(s)
- Yinghua Xu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Huajun Zheng
- Key Laboratory of Reproduction Regulation of NPFPC, Shanghai Institute of Planned Parenthood Research, IRD, Fudan UniversityShanghai, China; Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Zhangjiang Hi-Tech ParkShanghai, China
| | - Ying Zhang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Yuezhu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Zhangjiang Hi-Tech Park Shanghai, China
| | - Jinlong Zhang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Zhe Li
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Shenghui Cui
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Xiaofang Xin
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Qiang Ye
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca NY, USA
| | - Junzhi Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control Beijing, China
| |
Collapse
|
26
|
Miller MR, Miller KA, Bian J, James ME, Zhang S, Lynch M, Callery PS, Hettick JM, Cockburn A, Liu J, Li C, Crane BR, Charon NW. Spirochaete flagella hook proteins self-catalyse a lysinoalanine covalent crosslink for motility. Nat Microbiol 2016; 1:16134. [PMID: 27670115 PMCID: PMC5077173 DOI: 10.1038/nmicrobiol.2016.134] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/06/2016] [Indexed: 12/25/2022]
Abstract
Spirochaetes are bacteria responsible for several serious diseases, including Lyme disease (Borrelia burgdorferi), syphilis (Treponema pallidum) and leptospirosis (Leptospira interrogans), and contribute to periodontal diseases (Treponema denticola)(1). These spirochaetes employ an unusual form of flagella-based motility necessary for pathogenicity; indeed, spirochaete flagella (periplasmic flagella) reside and rotate within the periplasmic space(2-11). The universal joint or hook that links the rotary motor to the filament is composed of ∼120-130 FlgE proteins, which in spirochaetes form an unusually stable, high-molecular-weight complex(9,12-17). In other bacteria, the hook can be readily dissociated by treatments such as heat(18). In contrast, spirochaete hooks are resistant to these treatments, and several lines of evidence indicate that the high-molecular-weight complex is the consequence of covalent crosslinking(12,13,17). Here, we show that T. denticola FlgE self-catalyses an interpeptide crosslinking reaction between conserved lysine and cysteine, resulting in the formation of an unusual lysinoalanine adduct that polymerizes the hook subunits. Lysinoalanine crosslinks are not needed for flagellar assembly, but they are required for cell motility and hence infection. The self-catalytic nature of FlgE crosslinking has important implications for protein engineering, and its sensitivity to chemical inhibitors provides a new avenue for the development of antimicrobials targeting spirochaetes.
Collapse
Affiliation(s)
- Michael R. Miller
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| | - Kelly A. Miller
- Department of Microbiology, Immunology, and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| | - Jiang Bian
- Department of Oral Biology, State University of New York, Buffalo, NY, 14214
| | - Milinda E. James
- Department of Microbiology, Immunology, and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| | - Sheng Zhang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Michael Lynch
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Patrick S. Callery
- Department of Pharmaceutical Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| | - Justin M. Hettick
- National Institute for Occupational Safety and Health, 1095 Willowdale Road, Morgantown WV 26505
| | - Andrew Cockburn
- Department of Microbiology, Immunology, and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| | - Jun Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, 77030
| | - Chunhao Li
- Department of Oral Biology, State University of New York, Buffalo, NY, 14214
| | - Brian R. Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Nyles W. Charon
- Department of Microbiology, Immunology, and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506
| |
Collapse
|
27
|
Wunder EA, Figueira CP, Benaroudj N, Hu B, Tong BA, Trajtenberg F, Liu J, Reis MG, Charon NW, Buschiazzo A, Picardeau M, Ko AI. A novel flagellar sheath protein, FcpA, determines filament coiling, translational motility and virulence for the Leptospira spirochete. Mol Microbiol 2016; 101:457-70. [PMID: 27113476 DOI: 10.1111/mmi.13403] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2016] [Indexed: 12/28/2022]
Abstract
Leptospira are unique among bacteria based on their helical cell morphology with hook-shaped ends and the presence of periplasmic flagella (PF) with pronounced spontaneous supercoiling. The factors that provoke such supercoiling, as well as the role that PF coiling plays in generating the characteristic hook-end cell morphology and motility, have not been elucidated. We have now identified an abundant protein from the pathogen L. interrogans, exposed on the PF surface, and named it Flagellar-coiling protein A (FcpA). The gene encoding FcpA is highly conserved among Leptospira and was not found in other bacteria. fcpA(-) mutants, obtained from clinical isolates or by allelic exchange, had relatively straight, smaller-diameter PF, and were not able to produce translational motility. These mutants lost their ability to cause disease in the standard hamster model of leptospirosis. Complementation of fcpA restored the wild-type morphology, motility and virulence phenotypes. In summary, we identified a novel Leptospira 36-kDa protein, the main component of the spirochete's PF sheath, and a key determinant of the flagella's coiled structure. FcpA is essential for bacterial translational motility and to enable the spirochete to penetrate the host, traverse tissue barriers, disseminate to cause systemic infection and reach target organs.
Collapse
Affiliation(s)
- Elsio A Wunder
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, New Haven, CT, 06520, USA.,Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Bahia, 40296-710, Brazil
| | - Cláudio P Figueira
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Bahia, 40296-710, Brazil
| | - Nadia Benaroudj
- Institut Pasteur, Unit of Biology of Spirochetes, 75724 Paris cedex 15, France
| | - Bo Hu
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Brian A Tong
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Felipe Trajtenberg
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, 11400, Uruguay
| | - Jun Liu
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Mitermayer G Reis
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Bahia, 40296-710, Brazil
| | - Nyles W Charon
- Department of Microbiology and Immunology, West Virginia University, Morgantown, WV, 26506, USA
| | - Alejandro Buschiazzo
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, 11400, Uruguay.,Department of Structural Biology and Chemistry, Institute Pasteur, 75724 Paris cedex15, France
| | - Mathieu Picardeau
- Institut Pasteur, Unit of Biology of Spirochetes, 75724 Paris cedex 15, France
| | - Albert I Ko
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, New Haven, CT, 06520, USA.,Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Bahia, 40296-710, Brazil
| |
Collapse
|
28
|
Analysis of a Spontaneous Non-Motile and Avirulent Mutant Shows That FliM Is Required for Full Endoflagella Assembly in Leptospira interrogans. PLoS One 2016; 11:e0152916. [PMID: 27044038 PMCID: PMC4820103 DOI: 10.1371/journal.pone.0152916] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/21/2016] [Indexed: 11/19/2022] Open
Abstract
Pathogenic Leptospira strains are responsible for leptospirosis, a worldwide emerging zoonotic disease. These spirochetes are unique amongst bacteria because of their corkscrew-like cell morphology and their periplasmic flagella. Motility is reported as an important virulence determinant, probably favoring entry and dissemination of pathogenic Leptospira in the host. However, proteins constituting the periplasmic flagella and their role in cell shape, motility and virulence remain poorly described. In this study, we characterized a spontaneous L. interrogans mutant strain lacking motility, correlated with the loss of the characteristic hook-shaped ends, and virulence in the animal model. Whole genome sequencing allowed the identification of one nucleotide deletion in the fliM gene resulting in a premature stop codon, thereby preventing the production of flagellar motor switch protein FliM. Genetic complementation restored cell morphology, motility and virulence comparable to those of wild type cells. Analyses of purified periplasmic flagella revealed a defect in flagella assembly, resulting in shortened flagella compared to the wild type strain. This also correlated with a lower amount of major filament proteins FlaA and FlaB. Altogether, these findings demonstrate that FliM is required for full and correct assembly of the flagella which is essential for motility and virulence.
Collapse
|
29
|
Schons-Fonseca L, da Silva JB, Milanez JS, Domingos RH, Smith JL, Nakaya HI, Grossman AD, Ho PL, da Costa RMA. Analysis of LexA binding sites and transcriptomics in response to genotoxic stress in Leptospira interrogans. Nucleic Acids Res 2016; 44:1179-91. [PMID: 26762976 PMCID: PMC4756842 DOI: 10.1093/nar/gkv1536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 12/15/2015] [Accepted: 12/25/2015] [Indexed: 11/13/2022] Open
Abstract
We determined the effects of DNA damage caused by ultraviolet radiation on gene expression in Leptospira interrogans using DNA microarrays. These data were integrated with DNA binding in vivo of LexA1, a regulator of the DNA damage response, assessed by chromatin immunoprecipitation and massively parallel DNA sequencing (ChIP-seq). In response to DNA damage, Leptospira induced expression of genes involved in DNA metabolism, in mobile genetic elements and defective prophages. The DNA repair genes involved in removal of photo-damage (e.g. nucleotide excision repair uvrABC, recombinases recBCD and resolvases ruvABC) were not induced. Genes involved in various metabolic pathways were down regulated, including genes involved in cell growth, RNA metabolism and the tricarboxylic acid cycle. From ChIP-seq data, we observed 24 LexA1 binding sites located throughout chromosome 1 and one binding site in chromosome 2. Expression of many, but not all, genes near those sites was increased following DNA damage. Binding sites were found as far as 550 bp upstream from the start codon, or 1 kb into the coding sequence. Our findings indicate that there is a shift in gene expression following DNA damage that represses genes involved in cell growth and virulence, and induces genes involved in mutagenesis and recombination.
Collapse
Affiliation(s)
- Luciane Schons-Fonseca
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Josefa B da Silva
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Juliana S Milanez
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil
| | - Renan H Domingos
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Janet L Smith
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Helder I Nakaya
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil
| | - Alan D Grossman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Paulo L Ho
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Renata M A da Costa
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210580, Brazil
| |
Collapse
|
30
|
Fouts DE, Matthias MA, Adhikarla H, Adler B, Amorim-Santos L, Berg DE, Bulach D, Buschiazzo A, Chang YF, Galloway RL, Haake DA, Haft DH, Hartskeerl R, Ko AI, Levett PN, Matsunaga J, Mechaly AE, Monk JM, Nascimento ALT, Nelson KE, Palsson B, Peacock SJ, Picardeau M, Ricaldi JN, Thaipandungpanit J, Wunder EA, Yang XF, Zhang JJ, Vinetz JM. What Makes a Bacterial Species Pathogenic?:Comparative Genomic Analysis of the Genus Leptospira. PLoS Negl Trop Dis 2016; 10:e0004403. [PMID: 26890609 PMCID: PMC4758666 DOI: 10.1371/journal.pntd.0004403] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/03/2016] [Indexed: 12/20/2022] Open
Abstract
Leptospirosis, caused by spirochetes of the genus Leptospira, is a globally widespread, neglected and emerging zoonotic disease. While whole genome analysis of individual pathogenic, intermediately pathogenic and saprophytic Leptospira species has been reported, comprehensive cross-species genomic comparison of all known species of infectious and non-infectious Leptospira, with the goal of identifying genes related to pathogenesis and mammalian host adaptation, remains a key gap in the field. Infectious Leptospira, comprised of pathogenic and intermediately pathogenic Leptospira, evolutionarily diverged from non-infectious, saprophytic Leptospira, as demonstrated by the following computational biology analyses: 1) the definitive taxonomy and evolutionary relatedness among all known Leptospira species; 2) genomically-predicted metabolic reconstructions that indicate novel adaptation of infectious Leptospira to mammals, including sialic acid biosynthesis, pathogen-specific porphyrin metabolism and the first-time demonstration of cobalamin (B12) autotrophy as a bacterial virulence factor; 3) CRISPR/Cas systems demonstrated only to be present in pathogenic Leptospira, suggesting a potential mechanism for this clade's refractoriness to gene targeting; 4) finding Leptospira pathogen-specific specialized protein secretion systems; 5) novel virulence-related genes/gene families such as the Virulence Modifying (VM) (PF07598 paralogs) proteins and pathogen-specific adhesins; 6) discovery of novel, pathogen-specific protein modification and secretion mechanisms including unique lipoprotein signal peptide motifs, Sec-independent twin arginine protein secretion motifs, and the absence of certain canonical signal recognition particle proteins from all Leptospira; and 7) and demonstration of infectious Leptospira-specific signal-responsive gene expression, motility and chemotaxis systems. By identifying large scale changes in infectious (pathogenic and intermediately pathogenic) vs. non-infectious Leptospira, this work provides new insights into the evolution of a genus of bacterial pathogens. This work will be a comprehensive roadmap for understanding leptospirosis pathogenesis. More generally, it provides new insights into mechanisms by which bacterial pathogens adapt to mammalian hosts.
Collapse
Affiliation(s)
- Derrick E. Fouts
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Michael A. Matthias
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Haritha Adhikarla
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Ben Adler
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Department of Microbiology, Monash University, Clayton, Australia
| | - Luciane Amorim-Santos
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia, Brazil
| | - Douglas E. Berg
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Dieter Bulach
- Victorian Bioinformatics Consortium, Monash University, Clayton, Victoria, Australia
| | - Alejandro Buschiazzo
- Institut Pasteur de Montevideo, Laboratory of Molecular and Structural Microbiology, Montevideo, Uruguay
- Institut Pasteur, Department of Structural Biology and Chemistry, Paris, France
| | - Yung-Fu Chang
- Department of Population Medicine & Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Renee L. Galloway
- Centers for Disease Control and Prevention (DHHS, CDC, OID, NCEZID, DHCPP, BSPB), Atlanta, Georgia, United States of America
| | - David A. Haake
- VA Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Daniel H. Haft
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Rudy Hartskeerl
- WHO/FAO/OIE and National Collaborating Centre for Reference and Research on Leptospirosis, KIT Biomedical Research, Royal Tropical Institute (KIT), Amsterdam, The Netherlands
| | - Albert I. Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia, Brazil
| | - Paul N. Levett
- Government of Saskatchewan, Disease Control Laboratory Regina, Canada
| | - James Matsunaga
- VA Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ariel E. Mechaly
- Institut Pasteur de Montevideo, Laboratory of Molecular and Structural Microbiology, Montevideo, Uruguay
| | - Jonathan M. Monk
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
| | - Ana L. T. Nascimento
- Centro de Biotecnologia, Instituto Butantan, São Paulo, SP, Brazil
- Programa Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, São Paulo, SP, Brazil
| | - Karen E. Nelson
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Bernhard Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
| | - Sharon J. Peacock
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mathieu Picardeau
- Institut Pasteur, Biology of Spirochetes Unit, National Reference Centre and WHO Collaborating Center for Leptospirosis, Paris, France
| | - Jessica N. Ricaldi
- Instituto de Medicina Tropical Alexander von Humboldt; Facultad de Medicina Alberto Hurtado, Universidd Peruana Cayetano Heredia, Lima, Peru
| | | | - Elsio A. Wunder
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia, Brazil
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jun-Jie Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Joseph M. Vinetz
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Instituto de Medicina Tropical Alexander von Humboldt; Facultad de Medicina Alberto Hurtado, Universidd Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina “Alexander von Humboldt,” Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
31
|
Lehmann JS, Corey VC, Ricaldi JN, Vinetz JM, Winzeler EA, Matthias MA. Whole Genome Shotgun Sequencing Shows Selection on Leptospira Regulatory Proteins During in vitro Culture Attenuation. Am J Trop Med Hyg 2015; 94:302-313. [PMID: 26711524 PMCID: PMC4751964 DOI: 10.4269/ajtmh.15-0401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/08/2015] [Indexed: 12/29/2022] Open
Abstract
Leptospirosis is the most common zoonotic disease worldwide with an estimated 500,000 severe cases reported annually, and case fatality rates of 12–25%, due primarily to acute kidney and lung injuries. Despite its prevalence, the molecular mechanisms underlying leptospirosis pathogenesis remain poorly understood. To identify virulence-related genes in Leptospira interrogans, we delineated cumulative genome changes that occurred during serial in vitro passage of a highly virulent strain of L. interrogans serovar Lai into a nearly avirulent isogenic derivative. Comparison of protein coding and computationally predicted noncoding RNA (ncRNA) genes between these two polyclonal strains identified 15 nonsynonymous single nucleotide variant (nsSNV) alleles that increased in frequency and 19 that decreased, whereas no changes in allelic frequency were observed among the ncRNA genes. Some of the nsSNV alleles were in six genes shown previously to be transcriptionally upregulated during exposure to in vivo-like conditions. Five of these nsSNVs were in evolutionarily conserved positions in genes related to signal transduction and metabolism. Frequency changes of minor nsSNV alleles identified in this study likely contributed to the loss of virulence during serial in vitro culture. The identification of new virulence-associated genes should spur additional experimental inquiry into their potential role in Leptospira pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael A. Matthias
- *Address correspondence to Michael A. Matthias, Department of Medicine, Division of Infectious Diseases, School of Medicine, University of California, San Diego School of Medicine, 9500 Gilman Drive, BRF 2, Room 4A15, La Jolla, CA 92093-0760. E-mail:
| |
Collapse
|
32
|
Spirochetal motility and chemotaxis in the natural enzootic cycle and development of Lyme disease. Curr Opin Microbiol 2015; 28:106-13. [PMID: 26519910 DOI: 10.1016/j.mib.2015.09.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 11/21/2022]
Abstract
Two-thirds of all bacterial genomes sequenced to-date possess an organelle for locomotion, referred to as flagella, periplasmic flagella or type IV pili. These genomes may also contain a chemotaxis-signaling system which governs flagellar rotation, thus leading a coordinated function for motility. Motility and chemotaxis are often crucial for infection or disease process caused by pathogenic bacteria. Although motility-associated genes are well-characterized in some organisms, the highly orchestrated synthesis, regulation, and assembly of periplasmic flagella in spirochetes are just being delineated. Recent advances were fostered by development of unique genetic manipulations in spirochetes coupled with cutting-edge imaging techniques. These contemporary advances in understanding the role of spirochetal motility and chemotaxis in host persistence and disease development are highlighted in this review.
Collapse
|
33
|
Wolgemuth CW. Flagellar motility of the pathogenic spirochetes. Semin Cell Dev Biol 2015; 46:104-12. [PMID: 26481969 DOI: 10.1016/j.semcdb.2015.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 01/13/2023]
Abstract
Bacterial pathogens are often classified by their toxicity and invasiveness. The invasiveness of a given bacterium is determined by how capable the bacterium is at invading a broad range of tissues in its host. Of mammalian pathogens, some of the most invasive come from a group of bacteria known as the spirochetes, which cause diseases, such as syphilis, Lyme disease, relapsing fever and leptospirosis. Most of the spirochetes are characterized by their distinct shapes and unique motility. They are long, thin bacteria that can be shaped like flat-waves, helices, or have more irregular morphologies. Like many other bacteria, the spirochetes use long, helical appendages known as flagella to move; however, the spirochetes enclose their flagella in the periplasm, the narrow space between the inner and outer membranes. Rotation of the flagella in the periplasm causes the entire cell body to rotate and/or undulate. These deformations of the bacterium produce the force that drives the motility of these organisms, and it is this unique motility that likely allows these bacteria to be highly invasive in mammals. This review will describe the current state of knowledge on the motility and biophysics of these organisms and provide evidence on how this knowledge can inform our understanding of spirochetal diseases.
Collapse
Affiliation(s)
- Charles W Wolgemuth
- University of Connecticut Health Center, Department of Cell Biology and Center for Cell Analysis and Modeling, Farmington, CT 06030-3505, United States; University of Arizona, Department of Physics and Molecular and Cellular Biology, Tucson, AZ 85721, United States.
| |
Collapse
|
34
|
Eshghi A, Pappalardo E, Hester S, Thomas B, Pretre G, Picardeau M. Pathogenic Leptospira interrogans exoproteins are primarily involved in heterotrophic processes. Infect Immun 2015; 83:3061-73. [PMID: 25987703 PMCID: PMC4496612 DOI: 10.1128/iai.00427-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/11/2015] [Indexed: 12/19/2022] Open
Abstract
Leptospirosis is a life-threatening and emerging zoonotic disease with a worldwide annual occurrence of more than 1 million cases. Leptospirosis is caused by spirochetes belonging to the genus Leptospira. The mechanisms of disease manifestation in the host remain elusive, and the roles of leptospiral exoproteins in these processes have yet to be determined. Our aim in this study was to assess the composition and quantity of exoproteins of pathogenic Leptospira interrogans and to construe how these proteins contribute to disease pathogenesis. Label-free quantitative mass spectrometry of proteins obtained from Leptospira spirochetes cultured in vitro under conditions mimicking infection identified 325 exoproteins. The majority of these proteins are conserved in the nonpathogenic species Leptospira biflexa, and proteins involved in metabolism and energy-generating functions were overrepresented and displayed the highest relative abundance in culture supernatants. Conversely, proteins of unknown function, which represent the majority of pathogen-specific proteins (presumably involved in virulence mechanisms), were underrepresented. Characterization of various L. interrogans exoprotein mutants in the animal infection model revealed host mortality rates similar to those of hosts infected with wild-type L. interrogans. Collectively, these results indicate that pathogenic Leptospira exoproteins primarily function in heterotrophic processes (the processes by which organisms utilize organic substances as nutrient sources) to maintain the saprophytic lifestyle rather than the virulence of the bacteria. The underrepresentation of proteins homologous to known virulence factors, such as toxins and effectors in the exoproteome, also suggests that disease manifesting from Leptospira infection is likely caused by a combination of the primary and potentially moonlight functioning of exoproteins.
Collapse
Affiliation(s)
- Azad Eshghi
- Institut Pasteur, Biology of Spirochetes Unit, Paris, France
| | - Elisa Pappalardo
- University of Oxford, Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Svenja Hester
- University of Oxford, Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Benjamin Thomas
- University of Oxford, Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Gabriela Pretre
- Institut Pasteur, Biology of Spirochetes Unit, Paris, France
| | | |
Collapse
|
35
|
Zhu W, Wang J, Zhu Y, Tang B, Zhang Y, He P, Zhang Y, Liu B, Guo X, Zhao G, Qin J. Identification of three extra-chromosomal replicons in Leptospira pathogenic strain and development of new shuttle vectors. BMC Genomics 2015; 16:90. [PMID: 25887950 PMCID: PMC4338851 DOI: 10.1186/s12864-015-1321-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 02/04/2015] [Indexed: 12/20/2022] Open
Abstract
Background The genome of pathogenic Leptospira interrogans contains two chromosomes. Plasmids and prophages are known to play specific roles in gene transfer in bacteria and can potentially serve as efficient genetic tools in these organisms. Although plasmids and prophage remnants have recently been reported in Leptospira species, their characteristics and potential applications in leptospiral genetic transformation systems have not been fully evaluated. Results Three extrachromosomal replicons designated lcp1 (65,732 bp), lcp2 (56,757 bp), and lcp3 (54,986 bp) in the L. interrogans serovar Linhai strain 56609 were identified through whole genome sequencing. All three replicons were stable outside of the bacterial chromosomes. Phage particles were observed in the culture supernatant of 56609 after mitomycin C induction, and lcp3, which contained phage-related genes, was considered to be an inducible prophage. L. interrogans–Escherichia coli shuttle vectors, constructed with the predicted replication elements of single rep or rep combined with parAB loci from the three plasmids were shown to successfully transform into both saprophytic and pathogenic Leptospira species, suggesting an essential function for rep genes in supporting auto-replication of the plasmids. Additionally, a wide distribution of homologs of the three rep genes was identified in L. interrogans isolates, and correlation tests showed that the transformability of the shuttle vectors in L. interrogans isolates depended, to certain extent, on genetic compatibility between the rep sequences of both plasmid and host. Conclusions Three extrachromosomal replicons co-exist in L. interrogans, one of which we consider to be an inducible prophage. The vectors constructed with the rep genes of the three replicons successfully transformed into saprophytic and pathogenic Leptospira species alike, but this was partly dependent on genetic compatibility between the rep sequences of both plasmid and host. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1321-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Weinan Zhu
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Jin Wang
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yongzhang Zhu
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Biao Tang
- State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai, 200433, China.
| | - Yunyi Zhang
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Ping He
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Yan Zhang
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Boyu Liu
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Xiaokui Guo
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Guoping Zhao
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, China. .,State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai, 200433, China.
| | - Jinhong Qin
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
36
|
de Alvarenga Mudadu M, Carvalho V, Leclercq SY. Nonclassically secreted proteins as possible antigens for vaccine development: a reverse vaccinology approach. Appl Biochem Biotechnol 2015; 175:3360-70. [PMID: 25672322 DOI: 10.1007/s12010-015-1507-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/21/2015] [Indexed: 01/10/2023]
Abstract
Reverse vaccinology strategies have already been applied to a variety of microorganisms and have contributed significantly to vaccine development. However, most of the studies focused on an individual organism or on proteins with signature sequence motifs commonly found in known secreted proteins from bacteria. In this work, we applied a reverse vaccinology strategy based on conservation, virulence, and nonclassically surface exposure criterions to identify potential antigens in two microorganisms with significant degree of genomic plasticity among isolates (Streptococcus pneumoniae and Leptospira spp.), which imposes a major limitation to the production of a multistrain component vaccine. PSORTb 3.0.2 was run to predict the subcellular localization of the proteins. OrthoMCL was run to identify groups of the most conserved proteins between strains. Virulence prediction was done for the most conserved proteins, and SecretomeP was run to predict the nonclassically secreted proteins among the potential virulence factors. Based on the above criteria, we identified 37 proteins conserved between 16 genomes of S. pneumoniae and 12 proteins conserved between 5 leptospiral genomes as potential vaccine candidates.
Collapse
|
37
|
Abstract
The mechanisms of disease pathogenesis in leptospirosis are poorly defined. Recent developments in the application of genetic tools in the study of Leptospira have advanced our understanding by allowing the assessment of mutants in animal models. As a result, a small number of essential virulence factors have been identified, though most do not have a clearly defined function. Significant advances have also been made in the in vitro characterization of leptospiral interaction with host structures, including extracellular matrix proteins (such as laminin, elastin, fibronectin, collagens), proteins related to hemostasis (fibrinogen, plasmin), and soluble mediators of complement resistance (factor H, C4b-binding protein), although none of these in vitro findings has been translated to the host animal. Binding to host structures may permit colonization of the host, prevention of blood clotting may contribute to hemorrhage, while interaction with complement resistance mediators may contribute to survival in serum. While not a classical intracellular pathogen, the interaction of leptospires and phagocytic cells appears complex, with bacteria surviving uptake and promoting apoptosis; mutants relating to these processes (such as cell invasion and oxidative stress resistance) are attenuated in vivo. Another feature of leptospiral biology is the high degree of functional redundancy and the surprising lack of attenuation of mutants in what appear to be certain virulence factors, such as LipL32 and LigB. While many advances have been made, there remains a lack of understanding of how Leptospira causes tissue pathology. It is likely that leptospires have many novel pathogenesis mechanisms that are yet to be identified.
Collapse
|
38
|
Abstract
Recent advances in molecular genetics, such as the ability to construct defined mutants, have allowed the study of virulence factors and more generally the biology in Leptospira. However, pathogenic leptospires remain much less easily transformable than the saprophyte L. biflexa and further development and improvement of genetic tools are required. Here, we review tools that have been used to genetically manipulate Leptospira. We also describe the major advances achieved in both genomics and postgenomics technologies, including transcriptomics and proteomics.
Collapse
|
39
|
Komi KK, Ge YM, Xin XY, Ojcius DM, Sun D, Hu WL, Zhao X, Lin X, Yan J. RETRACTED: ChpK and MazF of the toxin-antitoxin modules are involved in the virulence of Leptospira interrogans during infection. Microbes Infect 2015; 17:34-47. [PMID: 25461800 DOI: 10.1016/j.micinf.2014.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 11/20/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the corresponding author and the editorial office of Microbes and Infection. An independent reviewer of the retraction request was also appointed given that one of the authors is the Editor-in- Chief. For figure 1C, Lanes 1 and 2 appear to share some unexpected similarities, except for the bottom band, which also appear to be the band of interest. Sections of Figure 2C appear similar to sections of Figure 5D of a paper that had already appeared in Molecular Microbiology, volume 83, issue 5 (2012) 1006-1023. https://doi.org/10.1111/j.1365-2958.2012.07985.x. In figure 3A, Flow cytograms share identical/similar patterns highlighted in various colours. Peculiarly, some of these patterns can be seen as horizontal rotations of others along the axis that separates different quadrants. (ie red green & purple). Moreover, some quadrants appear to have very high densities of events that are suprisingly limited by quadrant gates (most noticeably quadrants B2 from the second column of panels. Figure 5A-B it was found that there were duplicated bands were produced. Figures 5C and 5D, it was found that bands across each individual gel appear identical. One of the conditions of submission of a paper for publication is that authors declare explicitly that the paper has not been previously published and is not under consideration for publication elsewhere. Re-use of any data should be appropriately cited. As such this article represents a misuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process”.
Collapse
Affiliation(s)
- Komi Koukoura Komi
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China.
| | - Yu-Mei Ge
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Xiao-Yang Xin
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - David M Ojcius
- Health Sciences Research Institute and Molecular Cell Biology Department, University of California, Merced, CA 95343, USA.
| | - Dexter Sun
- New York Presbyterian Hospital & Hospital for Special Surgery, Weill Medical College, Cornell University, New York, NY, USA.
| | - Wei-Lin Hu
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Xin Zhao
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Xu'ai Lin
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
40
|
Abstract
The development of methods for the construction of defined mutants of pathogenic Leptospira has been a breakthrough in the study of leptospiral virulence. These methods have allowed the identification of genes essential for infection in animal models. This chapter describes methods for random transposon mutagenesis of pathogenic leptospires, identification of transposon insertion sites using direct sequencing from genomic DNA and a nested PCR utilizing degenerate oligonucleotides, and methods for testing mutant attenuation in the hamster model of infection.
Collapse
|
41
|
Abstract
Members of the family Leptospiraceae are thin, spiral, highly motile bacteria that are best visualized by darkfield microscopy. These characteristics are shared with other members of the Order Spirochaetales, but few additional parallels exist among spirochetes. This chapter describes basal features of Leptospira Leptospira that are central to survival and, in the case of pathogenic leptospiral species, intimately linked with pathogenesis, including its morphology, characteristic motility, and unusual metabolism. This chapter also describes the general methodology and critical requirements for in vitro cultivation and storage of Leptospira within a laboratory setting.
Collapse
Affiliation(s)
- Caroline E Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 3P6, Canada,
| |
Collapse
|
42
|
Pathogenesis of leptospirosis: Cellular and molecular aspects. Vet Microbiol 2014; 172:353-8. [DOI: 10.1016/j.vetmic.2014.06.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/09/2014] [Accepted: 06/14/2014] [Indexed: 11/23/2022]
|
43
|
Toma C, Murray GL, Nohara T, Mizuyama M, Koizumi N, Adler B, Suzuki T. Leptospiral outer membrane protein LMB216 is involved in enhancement of phagocytic uptake by macrophages. Cell Microbiol 2014; 16:1366-77. [DOI: 10.1111/cmi.12296] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/05/2014] [Accepted: 03/18/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Claudia Toma
- Department of Molecular Bacteriology and Immunology; Graduate School of Medicine; University of the Ryukyus; Okinawa 903-0215 Japan
| | - Gerald L. Murray
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics; Department of Microbiology; Monash University; Clayton Vic. 3800 Australia
| | - Toshitsugu Nohara
- Department of Molecular Bacteriology and Immunology; Graduate School of Medicine; University of the Ryukyus; Okinawa 903-0215 Japan
| | - Masaru Mizuyama
- Department of Molecular Bacteriology and Immunology; Graduate School of Medicine; University of the Ryukyus; Okinawa 903-0215 Japan
| | - Nobuo Koizumi
- Department of Bacteriology I; National Institute of Infectious Diseases; Tokyo 162-8640 Japan
| | - Ben Adler
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics; Department of Microbiology; Monash University; Clayton Vic. 3800 Australia
| | - Toshihiko Suzuki
- Department of Molecular Bacteriology and Immunology; Graduate School of Medicine; University of the Ryukyus; Okinawa 903-0215 Japan
| |
Collapse
|
44
|
A putative regulatory genetic locus modulates virulence in the pathogen Leptospira interrogans. Infect Immun 2014; 82:2542-52. [PMID: 24686063 DOI: 10.1128/iai.01803-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Limited research has been conducted on the role of transcriptional regulators in relation to virulence in Leptospira interrogans, the etiological agent of leptospirosis. Here, we identify an L. interrogans locus that encodes a sensor protein, an anti-sigma factor antagonist, and two genes encoding proteins of unknown function. Transposon insertion into the gene encoding the sensor protein led to dampened transcription of the other 3 genes in this locus. This lb139 insertion mutant (the lb139(-) mutant) displayed attenuated virulence in the hamster model of infection and reduced motility in vitro. Whole-transcriptome analyses using RNA sequencing revealed the downregulation of 115 genes and the upregulation of 28 genes, with an overrepresentation of gene products functioning in motility and signal transduction and numerous gene products with unknown functions, predicted to be localized to the extracellular space. Another significant finding encompassed suppressed expression of the majority of the genes previously demonstrated to be upregulated at physiological osmolarity, including the sphingomyelinase C precursor Sph2 and LigB. We provide insight into a possible requirement for transcriptional regulation as it relates to leptospiral virulence and suggest various biological processes that are affected due to the loss of native expression of this genetic locus.
Collapse
|
45
|
High-temperature protein G is an essential virulence factor of Leptospira interrogans. Infect Immun 2013; 82:1123-31. [PMID: 24366253 DOI: 10.1128/iai.01546-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Leptospira interrogans is a global zoonotic pathogen and is the causative agent of leptospirosis, an endemic disease of humans and animals worldwide. There is limited understanding of leptospiral pathogenesis; therefore, further elucidation of the mechanisms involved would aid in vaccine development and the prevention of infection. HtpG (high-temperature protein G) is the bacterial homolog to the highly conserved molecular chaperone Hsp90 and is important in the stress responses of many bacteria. The specific role of HtpG, especially in bacterial pathogenesis, remains largely unknown. Through the use of an L. interrogans htpG transposon insertion mutant, this study demonstrates that L. interrogans HtpG is essential for virulence in the hamster model of acute leptospirosis. Complementation of the htpG mutant completely restored virulence. Surprisingly, the htpG mutant did not appear to show sensitivity to heat or oxidative stress, phenotypes common in htpG mutants in other bacterial species. Furthermore, the mutant did not show increased sensitivity to serum complement, reduced survival within macrophages, or altered protein or lipopolysaccharide expression. The underlying cause for attenuation thus remains unknown, but HtpG is a novel leptospiral virulence factor and one of only a very small number identified to date.
Collapse
|
46
|
Kassegne K, Hu W, Ojcius DM, Sun D, Ge Y, Zhao J, Yang XF, Li L, Yan J. Identification of collagenase as a critical virulence factor for invasiveness and transmission of pathogenic Leptospira species. J Infect Dis 2013; 209:1105-15. [PMID: 24277745 DOI: 10.1093/infdis/jit659] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Leptospirosis is a global zoonotic disease. Transmission of Leptospira from animals to humans occurs through contact with water contaminated with leptospire-containing urine of infected animals. However, the molecular basis for the invasiveness of Leptospira and transmission of leptospirosis remains unknown. METHODS Activity of Leptospira interrogans strain Lai colA gene product (ColA) to hydrolyze different collagenic substrates was determined by spectrophotometry. Expression and secretion of ColA during infection were detected by reverse-transcription quantitative polymerase chain reaction and Western blot assay. The colA gene-deleted (ΔcolA) and colA gene-complemented (CΔcolA) mutants were generated to determine the roles of ColA in transcytosis in vitro and virulence in hamsters. RESULTS Recombinant or native ColA hydrolyzed all the tested substrates in which type III collagen was the favorite substrate with 2.16 mg/mL Km and 35.6 h(-)(1) Kcat values. Coincubation of the spirochete with HUVEC or HEK293 cells directly caused the significant elevation of ColA expression and secretion. Compared with wild-type strain, ΔcolA mutant displayed much-attenuated transcytosis through HEK293 and HUVEC monolayers, and less leptospires in blood, lung, liver, kidney and urine and 25-fold-decreased 50% lethal dose and milder histopathological injury in hamsters. CONCLUSIONS The product of colA gene is a collagenase as a crucial virulence factor in the invasiveness and transmission of L. interrogans.
Collapse
Affiliation(s)
- Kokouvi Kassegne
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lehmann JS, Fouts DE, Haft DH, Cannella AP, Ricaldi JN, Brinkac L, Harkins D, Durkin S, Sanka R, Sutton G, Moreno A, Vinetz JM, Matthias MA. Pathogenomic inference of virulence-associated genes in Leptospira interrogans. PLoS Negl Trop Dis 2013; 7:e2468. [PMID: 24098822 PMCID: PMC3789758 DOI: 10.1371/journal.pntd.0002468] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 08/23/2013] [Indexed: 11/18/2022] Open
Abstract
Leptospirosis is a globally important, neglected zoonotic infection caused by spirochetes of the genus Leptospira. Since genetic transformation remains technically limited for pathogenic Leptospira, a systems biology pathogenomic approach was used to infer leptospiral virulence genes by whole genome comparison of culture-attenuated Leptospira interrogans serovar Lai with its virulent, isogenic parent. Among the 11 pathogen-specific protein-coding genes in which non-synonymous mutations were found, a putative soluble adenylate cyclase with host cell cAMP-elevating activity, and two members of a previously unstudied ∼15 member paralogous gene family of unknown function were identified. This gene family was also uniquely found in the alpha-proteobacteria Bartonella bacilliformis and Bartonella australis that are geographically restricted to the Andes and Australia, respectively. How the pathogenic Leptospira and these two Bartonella species came to share this expanded gene family remains an evolutionary mystery. In vivo expression analyses demonstrated up-regulation of 10/11 Leptospira genes identified in the attenuation screen, and profound in vivo, tissue-specific up-regulation by members of the paralogous gene family, suggesting a direct role in virulence and host-pathogen interactions. The pathogenomic experimental design here is generalizable as a functional systems biology approach to studying bacterial pathogenesis and virulence and should encourage similar experimental studies of other pathogens.
Collapse
Affiliation(s)
- Jason S Lehmann
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Marcsisin RA, Bartpho T, Bulach DM, Srikram A, Sermswan RW, Adler B, Murray GL. Use of a high-throughput screen to identify Leptospira mutants unable to colonize the carrier host or cause disease in the acute model of infection. J Med Microbiol 2013; 62:1601-1608. [DOI: 10.1099/jmm.0.058586-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The molecular basis for leptospirosis infection and colonization remains poorly understood, with no efficient methods available for screening libraries of mutants for attenuation. We analysed the attenuation of leptospiral transposon mutants in vivo using a high-throughput method by infecting animals with pooled sets of transposon mutants. A total of 95 mutants was analysed by this method in the hamster model of acute infection, and one mutant was identified as attenuated (M1233, lb058 mutant). All virulence factors identified in Leptospira to date have been characterized in the acute model of infection, neglecting the carrier host. To address this, a BALB/c mouse colonization model was established. The lb058 mutant and two mutants defective in LPS synthesis were colonization deficient in the mouse model. By applying the high-throughput screening method, a further five colonization-deficient mutants were identified for the mouse model; these included two mutants in genes encoding proteins with a predicted role in iron uptake (LB191/HbpA and LB194). Two attenuated mutants had transposon insertions in either la0589 or la2786 (encoding proteins of unknown function). The final attenuated mutant had an unexpected deletion of genes la0969–la0975 at the point of transposon insertion. This is the first description of defined, colonization-deficient mutants in a carrier host for Leptospira. These mutants were either not attenuated or only weakly attenuated in the hamster model of acute leptospirosis, thus illustrating that different factors that may be required in the carrier and acute models of leptospiral infection. High-throughput screening can reduce the number of animals used in virulence studies and increase the capacity to screen mutants for attenuation, thereby enhancing the likelihood of detecting unique virulence factors. A comparison of virulence factors required in the carrier and acute models of infection will help to unravel colonization and dissemination mechanisms of leptospirosis.
Collapse
Affiliation(s)
- Renee A. Marcsisin
- Department of Microbiology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Thanatchaporn Bartpho
- Melioidosis Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Dieter M. Bulach
- Victorian Bioinformatics Consortium, Monash University, Clayton, Victoria 3800, Australia
| | - Amporn Srikram
- Section of Food Technology, Faculty of Natural Resources and Agro-Industry, Kasetsart University Chalermphrakiate Sakon Nakhon Province Campus, Sakon Nakhon 47000, Thailand
| | - Rasana W. Sermswan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ben Adler
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
- Victorian Bioinformatics Consortium, Monash University, Clayton, Victoria 3800, Australia
- Department of Microbiology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Gerald L. Murray
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
- Department of Microbiology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| |
Collapse
|
49
|
Leptospiral LruA is required for virulence and modulates an interaction with mammalian apolipoprotein AI. Infect Immun 2013; 81:3872-9. [PMID: 23918777 DOI: 10.1128/iai.01195-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Leptospirosis is a worldwide zoonosis caused by spirochetes of the genus Leptospira. While understanding of pathogenesis remains limited, the development of mutagenesis in Leptospira has provided a powerful tool for identifying novel virulence factors. LruA is a lipoprotein that has been implicated in leptospiral uveitis as a target of the immune response. In this study, two lruA mutants, M754 and M765, generated by transposon mutagenesis from Leptospira interrogans serovar Manilae, were characterized. In M754, the transposon inserted in the middle of lruA, resulting in no detectable expression of LruA. In M765, the transposon inserted toward the 3' end of the gene, resulting in expression of a truncated protein. LruA was demonstrated to be on the cell surface in M765 and the wild type (WT). M754, but not M765, was attenuated in a hamster model of acute infection. A search for differential binding to human serum proteins identified a serum protein of around 30 kDa bound to the wild type and the LruA deletion mutant (M754), but not to the LruA truncation mutant (M765). Two-dimensional separation of proteins from leptospiral cells incubated with guinea pig serum identified the 28-kDa apolipoprotein A-I (ApoA-I) as a major mammalian serum protein that binds Leptospira in vitro. Interestingly, M754 (with no detectable LruA) bound more ApoA-I than did the LruA-expressing strains Manilae wild type and M765. Our data thus identify LruA as a surface-exposed leptospiral virulence factor that contributes to leptospiral pathogenesis, possibly by modulating cellular interactions with serum protein ApoA-I.
Collapse
|
50
|
Leptospiral outer membrane protein LipL41 is not essential for acute leptospirosis but requires a small chaperone protein, lep, for stable expression. Infect Immun 2013; 81:2768-76. [PMID: 23690405 DOI: 10.1128/iai.00531-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leptospirosis is a worldwide zoonosis caused by pathogenic Leptospira spp., but knowledge of leptospiral pathogenesis remains limited. However, the development of mutagenesis systems has allowed the investigation of putative virulence factors and their involvement in leptospirosis. LipL41 is the third most abundant lipoprotein found in the outer membranes of pathogenic leptospires and has been considered a putative virulence factor. LipL41 is encoded on the large chromosome 28 bp upstream of a small open reading frame encoding a hypothetical protein of unknown function. This gene was named lep, for LipL41 expression partner. In this study, lipL41 was found to be cotranscribed with lep. Two transposon mutants were characterized: a lipL41 mutant and a lep mutant. In the lep mutant, LipL41 protein levels were reduced by approximately 90%. Lep was shown through cross-linking and coexpression experiments to bind to LipL41. Lep is proposed to be a molecular chaperone essential for the stable expression of LipL41. The roles of LipL41 and Lep in the pathogenesis of Leptospira interrogans were investigated; surprisingly, neither of these two unique proteins was essential for acute leptospirosis.
Collapse
|