51
|
D'Argenio V, Sarnataro D. Microbiome Influence in the Pathogenesis of Prion and Alzheimer's Diseases. Int J Mol Sci 2019; 20:E4704. [PMID: 31547531 PMCID: PMC6801937 DOI: 10.3390/ijms20194704] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 12/14/2022] Open
Abstract
Misfolded and abnormal β-sheets forms of wild-type proteins, such as cellular prion protein (PrPC) and amyloid beta (Aβ), are believed to be the vectors of neurodegenerative diseases, prion and Alzheimer's disease (AD), respectively. Increasing evidence highlights the "prion-like" seeding of protein aggregates as a mechanism for pathological spread in AD, tauopathy, as well as in other neurodegenerative diseases, such as Parkinson's. Mutations in both PrPC and Aβ precursor protein (APP), have been associated with the pathogenesis of these fatal disorders with clear evidence for their pathogenic significance. In addition, a critical role for the gut microbiota is emerging; indeed, as a consequence of gut-brain axis alterations, the gut microbiota has been involved in the regulation of Aβ production in AD and, through the microglial inflammation, in the amyloid fibril formation, in prion diseases. Here, we aim to review the role of microbiome ("the other human genome") alterations in AD and prion disease pathogenesis.
Collapse
Affiliation(s)
- Valeria D'Argenio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, via Pansini 5, 80131 Naples, Italy.
- CEINGE-Biotecnologie Avanzate, via G. Salvatore 486, 80145 Naples, Italy.
- Task Force on Microbiome Studies, University of Naples Federico II, 80131 Naples, Italy.
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, via Pansini 5, 80131 Naples, Italy.
- CEINGE-Biotecnologie Avanzate, via G. Salvatore 486, 80145 Naples, Italy.
| |
Collapse
|
52
|
Li B, He Y, Ma J, Huang P, Du J, Cao L, Wang Y, Xiao Q, Tang H, Chen S. Mild cognitive impairment has similar alterations as Alzheimer's disease in gut microbiota. Alzheimers Dement 2019; 15:1357-1366. [PMID: 31434623 DOI: 10.1016/j.jalz.2019.07.002] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/03/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Gut microbiota changes before the onset of Alzheimer's disease (AD) and the alterations could be detected in the stage of mild cognitive impairment (MCI). The findings might offer diagnostic biomarkers before the onset of dementia. BACKGROUND AD is the most common cause of dementia, and MCI is the predementia state. Recent studies suggest the alterations in the gut microbial communities associated with AD, whereas the microbiota in MCI before the onset of dementia has not been discovered and characterized in humans. NEW/UPDATED HYPOTHESIS We hypothesize that the dysbiosis happens in the MCI stage. Patients with AD and MCI have decreased microbial diversity, and changes in gut microbiota could be detected for early detection of AD. In our preliminary study, we identified differences between AD and normal controls in 11 genera from the feces and 11 genera from the blood. No difference in genera between AD and MCI was detected. Using the diagnostic model from fecal samples with all different genera input, 93% (28 in 30) of patients with MCI could be identified correctly. MAJOR CHALLENGES FOR THE HYPOTHESIS The diagnosis of MCI and AD in the study was based on symptoms and neuroimaging, and AD biomarkers should be included for precise diagnosis in further validating studies. Besides, as the microbiota changes longitudinally, their relationship with the progress of dementia needs to be studied in the prospective studies. LINKAGE TO OTHER MAJOR THEORIES Escherichia was observed increased at genus level in both fecal and blood samples from AD and MCI. For AD biomarker, postmortem brain tissue from patients with AD showed lipopolysaccharides and gram-negative Escherichia coli fragments colocalize with amyloid plaque. In this way, the amyloid pathogenesis for AD would be triggered during MCI by gut microbiota shifting. Besides, systemic inflammatory reactions caused by compounds secreted by bacteria may impair the blood-brain barrier and promote neuroinflammation and/or neurodegeneration. Furthermore, abnormal metabolites caused by microbial gene functions have an impact on neurodegeneration.
Collapse
Affiliation(s)
- Binyin Li
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yixi He
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jianfang Ma
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Pei Huang
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Juanjuan Du
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Li Cao
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yan Wang
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Qin Xiao
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Huidong Tang
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.
| | - Shengdi Chen
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.
| |
Collapse
|
53
|
Dutta SK, Verma S, Jain V, Surapaneni BK, Vinayek R, Phillips L, Nair PP. Parkinson's Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J Neurogastroenterol Motil 2019; 25:363-376. [PMID: 31327219 PMCID: PMC6657920 DOI: 10.5056/jnm19044] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/17/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022] Open
Abstract
The role of the microbiome in health and human disease has emerged at the forefront of medicine in the 21st century. Over the last 2 decades evidence has emerged to suggest that inflammation-derived oxidative damage and cytokine induced toxicity may play a significant role in the neuronal damage associated with Parkinson’s disease (PD). Presence of pro-inflammatory cytokines and T cell infiltration has been observed in the brain parenchyma of patients with PD. Furthermore, evidence for inflammatory changes has been reported in the enteric nervous system, the vagus nerve branches and glial cells. The presence of α-synuclein deposits in the post-mortem brain biopsy in patients with PD has further substantiated the role of inflammation in PD. It has been suggested that the α-synuclein misfolding might begin in the gut and spread “prion like” via the vagus nerve into lower brainstem and ultimately to the midbrain; this is known as the Braak hypothesis. It is noteworthy that the presence of gastrointestinal symptoms (constipation, dysphagia, and hypersalivation), altered gut microbiota and leaky gut have been observed in PD patients several years prior to the clinical onset of the disease. These clinical observations have been supported by in vitro studies in mice as well, demonstrating the role of genetic (α-synuclein overexpression) and environmental (gut dysbiosis) factors in the pathogenesis of PD. The restoration of the gut microbiome in patients with PD may alter the clinical progression of PD and this alteration can be accomplished by carefully designed studies using customized probiotics and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Sudhir K Dutta
- Sinai Hospital, Baltimore, MD, USA.,University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Padmanabhan P Nair
- Sinai Hospital, Baltimore, MD, USA.,Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.,NonInvasive Technologies LLC, Elkridge, MD, USA
| |
Collapse
|
54
|
Ambrosini YM, Borcherding D, Kanthasamy A, Kim HJ, Willette AA, Jergens A, Allenspach K, Mochel JP. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front Aging Neurosci 2019; 11:130. [PMID: 31275138 PMCID: PMC6591269 DOI: 10.3389/fnagi.2019.00130] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
Identifying appropriate animal models is critical in developing translatable in vitro and in vivo systems for therapeutic drug development and investigating disease pathophysiology. These animal models should have direct biological and translational relevance to the underlying disease they are supposed to mimic. Aging dogs not only naturally develop a cognitive decline in many aspects including learning and memory deficits, but they also exhibit human-like individual variability in the aging process. Neurodegenerative processes that can be observed in both human and canine brains include the progressive accumulation of β-amyloid (Aβ) found as diffuse plaques in the prefrontal cortex (PFC), including the gyrus proreus (i.e., medial orbital PFC), as well as the hippocampus and the cerebral vasculature. Tau pathology, a marker of neurodegeneration and dementia progression, was also found in canine hippocampal synapses. Various epidemiological data show that human patients with neurodegenerative diseases have concurrent intestinal lesions, and histopathological changes in the gastrointestinal (GI) tract occurs decades before neurodegenerative changes. Gut microbiome alterations have also been reported in many neurodegenerative diseases including Alzheimer's (AD) and Parkinson's diseases, as well as inflammatory central nervous system (CNS) diseases. Interestingly, the dog gut microbiome more closely resembles human gut microbiome in composition and functional overlap compared to rodent models. This article reviews the physiology of the gut-brain axis (GBA) and its involvement with neurodegenerative diseases in humans. Additionally, we outline the advantages and weaknesses of current in vitro and in vivo models and discuss future research directions investigating major human neurodegenerative diseases such as AD and Parkinson's diseases using dogs.
Collapse
Affiliation(s)
- Yoko M. Ambrosini
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Dana Borcherding
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Hyun Jung Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Auriel A. Willette
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Department of Food Science and Human Nutrition, College of Agriculture and Life Sciences, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
55
|
Cugini C, Shanmugam M, Landge N, Ramasubbu N. The Role of Exopolysaccharides in Oral Biofilms. J Dent Res 2019; 98:739-745. [PMID: 31009580 DOI: 10.1177/0022034519845001] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The oral cavity contains a rich consortium of exopolysaccharide-producing microbes. These extracellular polysaccharides comprise a major component of the oral biofilm. Together with extracellular proteins, DNA, and lipids, they form the biofilm matrix, which contributes to bacterial colonization, biofilm formation and maintenance, and pathogenesis. While a number of oral microbes have been studied in detail with regard to biofilm formation and pathogenesis, the exopolysaccharides have been well characterized for only select organisms, namely Streptococcus mutans and Aggregatibacter actinomycetemcomitans. Studies on the exopolysaccharides of other oral organisms, however, are in their infancy. In this review, we present the current research on exopolysaccharides of oral microbes regarding their biosynthesis, regulation, contributions to biofilm formation and stability of the matrix, and immune evasion. In addition, insight into the role of exopolysaccharides in biofilms is highlighted through the evaluation of emerging techniques such as pH probing of biofilm colonies, solid-state nuclear magnetic resonance for macromolecular interactions within biofilms, and super-resolution microscopy analysis of biofilm development. Finally, exopolysaccharide as a potential nutrient source for species within a biofilm is discussed.
Collapse
Affiliation(s)
- C Cugini
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - M Shanmugam
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - N Landge
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - N Ramasubbu
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
56
|
Chen D, Cao Y, Yu L, Tao Y, Zhou Y, Zhi Q, Lin H. Characteristics and influencing factors of amyloid fibers in S. mutans biofilm. AMB Express 2019; 9:31. [PMID: 30820691 PMCID: PMC6395465 DOI: 10.1186/s13568-019-0753-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
There are signs that amyloid fibers exist in Streptococcus mutans biofilm recently. However, the characteristics of amyloid fibers and fibrillation influencing factors are unknown. In this study, we firstly used transmission electron microscopy (TEM) and atomic force microscopy (AFM) to observe the morphology of amyloid fibers in S. mutans. Then the extracted amyloid fibers from biofilm were studied for their characteristics. Further, the influencing factors, PH, temperature and eDNA, were investigated. Results showed there were mainly two morphologies of amyloid fibers in S. mutans, different in width. Amyloid fibers inhibitor-EGCG obviously destroyed biofilm at different stages, which is dose-dependent. The amount of amyloid fibers positively correlated with biofilm biomass in clinical isolates. Acidic pH and high temperature obviously accelerated amyloid fibrillation. During amyloid fibrillation, amyloid growth morphologies were observed by TEM and results showed two growth morphologies. Amyloid fibers formed complex with eDNA, which we call (a)eDNA. The molecular weight of (a)eDNA was similar to genomic DNA, greatly larger than that of eDNA in matrix. Combined use of DNase I and EGCG was more efficiently in inhibiting amyloid fibers and biofilm biomass. In conclusion, amyloid fibers are the crucial structures for S. mutans biofilm formation, showing two types of morphology. Acidic pH and temperature can obviously accelerate amyloid fibrillation. Amyloid fibers form complex with (a)eDNA and combined use of DNase and amyloid fiber inhibitor is more efficiently in inhibiting S. mutans biofilm formation.
Collapse
|
57
|
Lemos JA, Palmer SR, Zeng L, Wen ZT, Kajfasz JK, Freires IA, Abranches J, Brady LJ. The Biology of Streptococcus mutans. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0051-2018. [PMID: 30657107 PMCID: PMC6615571 DOI: 10.1128/microbiolspec.gpp3-0051-2018] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
As a major etiological agent of human dental caries, Streptococcus mutans resides primarily in biofilms that form on the tooth surfaces, also known as dental plaque. In addition to caries, S. mutans is responsible for cases of infective endocarditis with a subset of strains being indirectly implicated with the onset of additional extraoral pathologies. During the past 4 decades, functional studies of S. mutans have focused on understanding the molecular mechanisms the organism employs to form robust biofilms on tooth surfaces, to rapidly metabolize a wide variety of carbohydrates obtained from the host diet, and to survive numerous (and frequent) environmental challenges encountered in oral biofilms. In these areas of research, S. mutans has served as a model organism for ground-breaking new discoveries that have, at times, challenged long-standing dogmas based on bacterial paradigms such as Escherichia coli and Bacillus subtilis. In addition to sections dedicated to carbohydrate metabolism, biofilm formation, and stress responses, this article discusses newer developments in S. mutans biology research, namely, how S. mutans interspecies and cross-kingdom interactions dictate the development and pathogenic potential of oral biofilms and how next-generation sequencing technologies have led to a much better understanding of the physiology and diversity of S. mutans as a species.
Collapse
Affiliation(s)
- J A Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - S R Palmer
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH 43210
| | - L Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - Z T Wen
- Department of Comprehensive Dentistry and Biomaterials and Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - J K Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - I A Freires
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - J Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - L J Brady
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| |
Collapse
|
58
|
Bacterial Amyloids: Biogenesis and Biomaterials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1174:113-159. [DOI: 10.1007/978-981-13-9791-2_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
59
|
Jansens KJA, Rombouts I, Grootaert C, Brijs K, Van Camp J, Van der Meeren P, Rousseau F, Schymkowitz J, Delcour JA. Rational Design of Amyloid-Like Fibrillary Structures for Tailoring Food Protein Techno-Functionality and Their Potential Health Implications. Compr Rev Food Sci Food Saf 2018; 18:84-105. [PMID: 33337021 DOI: 10.1111/1541-4337.12404] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/30/2022]
Abstract
To control and enhance protein functionality is a major challenge for food scientists. In this context, research on food protein fibril formation, especially amyloid fibril formation, holds much promise. We here first provide a concise overview of conditions, which affect amyloid formation in food proteins. Particular attention is directed towards amyloid core regions because these sequences promote ordered aggregation. Better understanding of this process will be key to tailor the fibril formation process. Especially seeding, that is, adding preformed protein fibrils to protein solutions to accelerate fibril formation holds promise to tailor aggregation and fibril techno-functionality. Some studies have already indicated that food protein fibrillation indeed improves their techno-functionality. However, much more research is necessary to establish whether protein fibrils are useful in complex food systems and whether and to what extent they resist food processing unit operations. In this review the effect of amyloid formation on gelation, interfacial properties, foaming, and emulsification is discussed. Despite their prevalent role as functional structures, amyloids also receive a lot of attention due to their association with protein deposition diseases, prompting us to thoroughly investigate the potential health impact of amyloid-like aggregates in food. A literature review on the effect of the different stages of the human digestive process on amyloid toxicity leads us to conclude that food-derived amyloid fibrils (even those with potential pathogenic properties) very likely have minimal impact on human health. Nevertheless, prior to wide-spread application of the technology, it is highly advisable to further verify the lack of toxicity of food-derived amyloid fibrils.
Collapse
Affiliation(s)
- Koen J A Jansens
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001, Leuven, Belgium
| | - Ine Rombouts
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001, Leuven, Belgium
| | - Charlotte Grootaert
- Laboratory of Food Chemistry and Human Nutrition, Ghent Univ., Coupure Links 653, B-9000, Ghent, Belgium
| | - Kristof Brijs
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001, Leuven, Belgium
| | - John Van Camp
- Laboratory of Food Chemistry and Human Nutrition, Ghent Univ., Coupure Links 653, B-9000, Ghent, Belgium
| | - Paul Van der Meeren
- Particle and Interfacial Technology Group, Ghent Univ., Coupure Links 653, B- 9000, Ghent, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB, B-3000 Leuven, Belgium. Authors Rousseau and Schymkowitz are also with Dept. of Cellular and Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB, B-3000 Leuven, Belgium. Authors Rousseau and Schymkowitz are also with Dept. of Cellular and Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
| | - Jan A Delcour
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001, Leuven, Belgium
| |
Collapse
|
60
|
Paranjapye N, Daggett V. De Novo Designed α-Sheet Peptides Inhibit Functional Amyloid Formation of Streptococcus mutans Biofilms. J Mol Biol 2018; 430:3764-3773. [PMID: 30006266 PMCID: PMC6168415 DOI: 10.1016/j.jmb.2018.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/29/2018] [Accepted: 07/02/2018] [Indexed: 12/11/2022]
Abstract
Streptococcus mutans is a bacterial species that predominates in the oral microbiome. S. mutans binds to the tooth surface, metabolizes sugars and produces acid, leading to cavity formation. S. mutans can also cause infectious endocarditis. Recent evidence suggests that S. mutans biofilms contain amyloid fibrils. Amyloids are insoluble fibrillar protein aggregates, and bacteria use functional amyloids to improve robustness of their biofilms. While the functional amyloids in bacteria such as Escherichia coli and Staphylococcus aureus have been heavily investigated, little is known about the mechanism of S. mutans amyloid formation. Previous results from our laboratory with the amyloidogenic proteins and peptides from the aforementioned bacteria and other mammalian amyloid systems suggest that amyloid formation progresses via an intermediate that adopts a unique secondary structure-α-sheet. De novo designed peptides with alternating l- and d-amino acid also adopt an α-sheet secondary structure and inhibit amyloid formation by binding to soluble oligomeric species during amyloidogenesis. Inhibition of fibrillization by α-sheet peptides suggests the presence of α-sheet during amyloid formation. To investigate the mechanism of functional amyloid formation in S. mutans, α-sheet peptides were compared to epigallocatechin gallate for their ability to inhibit fibril formation in S. mutans. Inhibition was demonstrated in a biofilm plate assay and on hydroxyapatite surfaces both in S. mutans alone and in bacteria from human saliva. The observed inhibition suggests that an α-sheet mediated mechanism may be operative during functional amyloid formation.
Collapse
Affiliation(s)
- Natasha Paranjapye
- Department of Bioengineering, University of Washington, Box 355013, Seattle, WA 98195-5013, USA
| | - Valerie Daggett
- Department of Bioengineering, University of Washington, Box 355013, Seattle, WA 98195-5013, USA.
| |
Collapse
|
61
|
Erskine E, MacPhee CE, Stanley-Wall NR. Functional Amyloid and Other Protein Fibers in the Biofilm Matrix. J Mol Biol 2018; 430:3642-3656. [PMID: 30098341 PMCID: PMC6173796 DOI: 10.1016/j.jmb.2018.07.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
Biofilms are ubiquitous in the natural and man-made environment. They are defined as microbes that are encapsulated in an extracellular, self-produced, biofilm matrix. Growing evidence from the genetic and biochemical analysis of single species biofilms has linked the presence of fibrous proteins to a functional biofilm matrix. Some of these fibers have been described as functional amyloid or amyloid-like fibers. Here we provide an overview of the biophysical and biological data for a wide range of protein fibers found in the biofilm matrix of Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Elliot Erskine
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Cait E MacPhee
- James Clerk Maxwell Building, School of Physics, University of Edinburgh, The Kings Buildings, Mayfield Road, Edinburgh, EH9 3JZ, UK.
| | - Nicola R Stanley-Wall
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| |
Collapse
|
62
|
Wang Y, Jiang J, Gao Y, Sun Y, Dai J, Wu Y, Qu D, Ma G, Fang X. Staphylococcus epidermidis small basic protein (Sbp) forms amyloid fibrils, consistent with its function as a scaffolding protein in biofilms. J Biol Chem 2018; 293:14296-14311. [PMID: 30049797 PMCID: PMC6139570 DOI: 10.1074/jbc.ra118.002448] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Biofilms are communities of microbes embedded in a microbial extracellular matrix. Their formation is considered the main virulence mechanism enabling the opportunistic bacterial pathogen Staphylococcus epidermidis to cause devastating nosocomial, implant-associated infections. Biofilms often contain proteins, and an 18-kDa protein called small basic protein (Sbp) recently was discovered in the S. epidermidis biofilm matrix and may serve as a scaffolding protein in both polysaccharide intercellular adhesin (PIA)-dependent and accumulation-associated protein (Aap)-dependent biofilm formations. In Aap-mediated biofilm formation, Sbp colocalizes with Domain-B of Aap, implying that Sbp directly interacts with Aap's Domain-B. However, the structure of Sbp and its interaction with Aap, as well as the molecular mechanism underlying Sbp's roles in biofilm formation, are incompletely understood. In this work, we used small-angle X-ray scattering (SAXS), NMR, analytical size-exclusion chromatography, and isothermal titration calorimetry analyses to determine the Sbp structure and characterize its interaction with Aap's Domain-B. We found that Sbp is monomeric and partially folded in solution, and, unexpectedly, we observed no direct interactions between Sbp and Aap Domain-B. Instead, we noted that Sbp forms amyloid fibrils both in vitro and in vivo Atomic force, transmission electron, and confocal fluorescence microscopy methods confirmed the formation of Sbp amyloid fibrils and revealed their morphology. Taken together, the Sbp amyloid fibril structures identified here may account for Sbp's role as a scaffolding protein in the S. epidermidis biofilm matrix.
Collapse
Affiliation(s)
- Yan Wang
- From the Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingbo Jiang
- From the Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yachao Gao
- From the Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yang Sun
- From the Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianfeng Dai
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education and Ministry of Public Health, Department of Medical Microbiology and Parasitology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, China, and
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education and Ministry of Public Health, Department of Medical Microbiology and Parasitology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, China, and
| | - Gang Ma
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding 071002, China
| | - Xianyang Fang
- From the Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China,
| |
Collapse
|
63
|
Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: The role of the intestinal microbiota in neurodegenerative diseases. Neurochem Int 2018; 120:149-163. [PMID: 30114473 DOI: 10.1016/j.neuint.2018.08.005] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023]
Abstract
The number of bacterial cells living within the human body is approximately equal to, or greater than, the total number of human cells. This dynamic population of microorganisms, termed the human microbiota, resides mainly within the gastrointestinal tract. It is widely accepted that highly diverse and stable microbiota promote overall human health. Colonization of the gut with maladaptive and pathogenic microbiota, a state also known as dysbiosis, is associated with a variety of peripheral diseases ranging from type 2 diabetes mellitus to cardiovascular and inflammatory bowel disease. More recently, microbial dysbiosis has been associated with a number of brain pathologies, including autism spectrum disorder, Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), suggesting a direct or indirect communication between intestinal bacteria and the central nervous system (CNS). In this review, we illustrate two pathways implicated in the crosstalk between gut microbiota and CNS involving 1) the vagus nerve and 2) transmission of signaling molecules through the circulatory system and across the blood-brain barrier (BBB). We summarize the available evidence of the specific changes in the intestinal microbiota, as well as microorganism-induced modifications to intestinal and BBB permeability, which have been linked to several neurodegenerative disorders including ALS, AD, and PD. Even though each of these diseases arises from unique pathogenetic mechanisms, all are characterized, at least in part, by chronic neuroinflammation. We provide an interpretation for the substantial evidence that healthy intestinal microbiota have the ability to positively regulate the neuroimmune responses in the CNS. Even though the evidence is mainly associative, it has been suggested that bacterial dysbiosis could contribute to an adverse neuroinflammatory state leading to increased risk of neurodegenerative diseases. Thus, developing strategies for regulating and maintaining healthy intestinal microbiota could be a valid approach for lowering individual risk and prevalence of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Deanna Lynn Gibson
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, Canada
| |
Collapse
|
64
|
Van Gerven N, Van der Verren SE, Reiter DM, Remaut H. The Role of Functional Amyloids in Bacterial Virulence. J Mol Biol 2018; 430:3657-3684. [PMID: 30009771 PMCID: PMC6173799 DOI: 10.1016/j.jmb.2018.07.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/14/2022]
Abstract
Amyloid fibrils are best known as a product of human and animal protein misfolding disorders, where amyloid formation is associated with cytotoxicity and disease. It is now evident that for some proteins, the amyloid state constitutes the native structure and serves a functional role. These functional amyloids are proving widespread in bacteria and fungi, fulfilling diverse functions as structural components in biofilms or spore coats, as toxins and surface-active fibers, as epigenetic material, peptide reservoirs or adhesins mediating binding to and internalization into host cells. In this review, we will focus on the role of functional amyloids in bacterial pathogenesis. The role of functional amyloids as virulence factor is diverse but mostly indirect. Nevertheless, functional amyloid pathways deserve consideration for the acute and long-term effects of the infectious disease process and may form valid antimicrobial targets. Functional amyloids are widespread in bacteria, pathogenic and non-pathogenic. Bacterial biofilms most commonly function as structural support in the extracellular matrix of biofilms or spore coats, and in cell–cell and cell-surface adherence. The amyloid state can be the sole structured and functional state, or can be facultative, as a secondary state to folded monomeric subunits. Bacterial amyloids can enhance virulence by increasing persistence, cell adherence and invasion, intracellular survival, and pathogen spread by increased environmental survival. Bacterial amyloids may indirectly inflict disease by triggering inflammation, contact phase activation and possibly induce or aggravate human pathological aggregation disorders.
Collapse
Affiliation(s)
- Nani Van Gerven
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Sander E Van der Verren
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Dirk M Reiter
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Han Remaut
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
65
|
Fünfhaus A, Göbel J, Ebeling J, Knispel H, Garcia-Gonzalez E, Genersch E. Swarming motility and biofilm formation of Paenibacillus larvae, the etiological agent of American Foulbrood of honey bees (Apis mellifera). Sci Rep 2018; 8:8840. [PMID: 29892084 PMCID: PMC5995878 DOI: 10.1038/s41598-018-27193-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022] Open
Abstract
American Foulbrood is a worldwide distributed, fatal disease of the brood of the Western honey bee (Apis mellifera). The causative agent of this fatal brood disease is the Gram-positive, spore-forming bacterium Paenibacillus larvae, which can be classified into four different genotypes (ERIC I-IV), with ERIC I and II being the ones isolated from contemporary AFB outbreaks. P. larvae is a peritrichously flagellated bacterium and, hence, we hypothesized that P. larvae is capable of coordinated and cooperative multicellular behaviors like swarming motility and biofilm formation. In order to analyze these behaviors of P. larvae, we firstly established appropriate functional assays. Using these assays we demonstrated that P. larvae ERIC II, but not P. larvae ERIC I, was capable of swarming. Swarming motility was hampered in a P. larvae ERIC II-mutant lacking production of paenilarvin, an iturin-like lipopeptide exclusively expressed by this genotype. Both genotypes were able to form free floating biofilm aggregates loosely attached to the walls of the culture wells. Visualizing the biofilms by Congo red and thioflavin S staining suggested structural differences between the biofilms formed. Biofilm formation was shown to be independent from paenilarvin production because the paenilarvin deficient mutant was comparably able to form a biofilm.
Collapse
Affiliation(s)
- Anne Fünfhaus
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany
| | - Josefine Göbel
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany
| | - Julia Ebeling
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany
| | - Henriette Knispel
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany
| | - Eva Garcia-Gonzalez
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany
| | - Elke Genersch
- Institute for Bee Research, Department of Molecular Microbiology and Bee Diseases, Hohen Neuendorf, Germany.
- Freie Universität Berlin, Fachbereich Veterinärmedizin, Institut für Mikrobiologie und Tierseuchen, Berlin, Germany.
| |
Collapse
|
66
|
Abstract
The name human contact system is related to its mode of action, as "contact" with artificial negatively charged surfaces triggers its activation. Today, it is generally believed that the contact system is an inflammatory response mechanism not only against artificial material but also against misfolded proteins and foreign organisms. Upon activation, the contact system is involved in at least two distinct (patho)physiologic processes:i. the trigger of the intrinsic coagulation via factor XI and ii. the cleavage of high molecular weight kininogen with release of bradykinin and antimicrobial peptides (AMPs). Bradykinin is involved in the regulation of inflammatory processes, vascular permeability, and blood pressure. Due to the release of AMPs, the contact system is regarded as a branch of the innate immune defense against microorganisms. There is an increasing list of pathogens that interact with contact factors, in addition to bacteria also fungi and viruses bind and activate the system. In spite of that, pathogens have developed their own mechanisms to activate the contact system, resulting in manipulation of this host immune response. In this up-to-date review, we summarize present research on the interaction of pathogens with the human contact system, focusing particularly on bacterial and viral mechanisms that trigger inflammation via contact system activation.
Collapse
Affiliation(s)
- Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| | - Juliane Köhler
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
67
|
Peterson CP, Sauer C, Chatfield CH. The Extracellular Polymeric Substances of Legionella pneumophila Biofilms Contain Amyloid Structures. Curr Microbiol 2018; 75:736-744. [PMID: 29468303 DOI: 10.1007/s00284-018-1440-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
Human infection by bacteria of the genus Legionella most often result in the pneumonia known as Legionnaires Disease. Legionella is found as a resident of adherent biofilms in man-made water systems. Disinfection efforts to prevent Legionella infections require a better understanding of the structures that promote Legionella surface attachment and biofilm colonization. Various enzymatic treatments, including multiple carbohydrate-targeting mixtures, failed to disrupt Legionella biofilms, despite the presence of carbohydrates in the biofilms as shown by biochemical methods and concanavalin-A lectin staining. Moreover, Legionella biofilms contained amyloids as detected by three microscopic staining methods (congo red, thioflavin T, and the amyloid-specific antibody WO2). Amyloid structures were seen in biofilms of both L. pneumophila and L. longbeachae, the two Legionella species most associated with human infection. Inhibition of amyloid assembly by congo red and thioflavin T limited both self-aggregation and surface attachment of L. pneumophila, indicating that functional amyloid structures have a key role in initial biofilm formation by these pathogenic bacteria.
Collapse
Affiliation(s)
- Casey P Peterson
- Department of Biological Sciences, SUNY Cortland, Cortland, NY, USA
| | - Cassidy Sauer
- Department of Biological Sciences, SUNY Cortland, Cortland, NY, USA
| | | |
Collapse
|
68
|
Desvaux M, Candela T, Serror P. Surfaceome and Proteosurfaceome in Parietal Monoderm Bacteria: Focus on Protein Cell-Surface Display. Front Microbiol 2018; 9:100. [PMID: 29491848 PMCID: PMC5817068 DOI: 10.3389/fmicb.2018.00100] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
The cell envelope of parietal monoderm bacteria (archetypal Gram-positive bacteria) is formed of a cytoplasmic membrane (CM) and a cell wall (CW). While the CM is composed of phospholipids, the CW is composed at least of peptidoglycan (PG) covalently linked to other biopolymers, such as teichoic acids, polysaccharides, and/or polyglutamate. Considering the CW is a porous structure with low selective permeability contrary to the CM, the bacterial cell surface hugs the molecular figure of the CW components as a well of the external side of the CM. While the surfaceome corresponds to the totality of the molecules found at the bacterial cell surface, the proteinaceous complement of the surfaceome is the proteosurfaceome. Once translocated across the CM, secreted proteins can either be released in the extracellular milieu or exposed at the cell surface by associating to the CM or the CW. Following the gene ontology (GO) for cellular components, cell-surface proteins at the CM can either be integral (GO: 0031226), i.e., the integral membrane proteins, or anchored to the membrane (GO: 0046658), i.e., the lipoproteins. At the CW (GO: 0009275), cell-surface proteins can be covalently bound, i.e., the LPXTG-proteins, or bound through weak interactions to the PG or wall polysaccharides, i.e., the cell wall binding proteins. Besides monopolypeptides, some proteins can associate to each other to form supramolecular protein structures of high molecular weight, namely the S-layer, pili, flagella, and cellulosomes. After reviewing the cell envelope components and the different molecular mechanisms involved in protein attachment to the cell envelope, perspectives in investigating the proteosurfaceome in parietal monoderm bacteria are further discussed.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRA, UMR454 MEDiS, Clermont-Ferrand, France
| | - Thomas Candela
- EA4043 Unité Bactéries Pathogènes et Santé, Châtenay-Malabry, France
| | - Pascale Serror
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
69
|
Upadhyay A, Mishra A. Amyloids of multiple species: are they helpful in survival? Biol Rev Camb Philos Soc 2018; 93:1363-1386. [DOI: 10.1111/brv.12399] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 01/13/2018] [Accepted: 01/18/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| |
Collapse
|
70
|
Abstract
It has become apparent that the intestinal microbiota orchestrates important aspects of our metabolism, immunity, and development. Recent work has demonstrated that the microbiota also influences brain function in healthy and diseased individuals. Of great interest are reports that intestinal bacteria play a role in the pathogenic cascade of both Parkinson and Alzheimer diseases. These neurodegenerative disorders both involve misfolding of endogenous proteins that spreads from one region of the body to another in a manner analogous to prions. The mechanisms of how the microbiota influences or is correlated with disease require elaboration. Microbial proteins or metabolites may influence neurodegeneration through the promotion of amyloid formation by human proteins or by enhancing inflammatory responses to endogenous neuronal amyloids. We review the current knowledge concerning bacterial amyloids and their potential to influence cerebral amyloid aggregation and neuroinflammation. We propose the term “mapranosis” to describe the process of microbiota-associated proteopathy and neuroinflammation. The study of amyloid proteins made by the microbiota and their influence on health and disease is in its infancy. This is a promising area for therapeutic intervention because there are many ways to alter our microbial partners and their products, including amyloid proteins.
Collapse
Affiliation(s)
- Robert P. Friedland
- Department of Neurology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| | - Matthew R. Chapman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
71
|
Amyloid-Like β-Aggregates as Force-Sensitive Switches in Fungal Biofilms and Infections. Microbiol Mol Biol Rev 2017; 82:82/1/e00035-17. [PMID: 29187516 DOI: 10.1128/mmbr.00035-17] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cellular aggregation is an essential step in the formation of biofilms, which promote fungal survival and persistence in hosts. In many of the known yeast cell adhesion proteins, there are amino acid sequences predicted to form amyloid-like β-aggregates. These sequences mediate amyloid formation in vitro. In vivo, these sequences mediate a phase transition from a disordered state to a partially ordered state to create patches of adhesins on the cell surface. These β-aggregated protein patches are called adhesin nanodomains, and their presence greatly increases and strengthens cell-cell interactions in fungal cell aggregation. Nanodomain formation is slow (with molecular response in minutes and the consequences being evident for hours), and strong interactions lead to enhanced biofilm formation. Unique among functional amyloids, fungal adhesin β-aggregation can be triggered by the application of physical shear force, leading to cellular responses to flow-induced stress and the formation of robust biofilms that persist under flow. Bioinformatics analysis suggests that this phenomenon may be widespread. Analysis of fungal abscesses shows the presence of surface amyloids in situ, a finding which supports the idea that phase changes to an amyloid-like state occur in vivo. The amyloid-coated fungi bind the damage-associated molecular pattern receptor serum amyloid P component, and there may be a consequential modulation of innate immune responses to the fungi. Structural data now suggest mechanisms for the force-mediated induction of the phase change. We summarize and discuss evidence that the sequences function as triggers for protein aggregation and subsequent cellular aggregation, both in vitro and in vivo.
Collapse
|
72
|
Pritchard AB, Crean S, Olsen I, Singhrao SK. Periodontitis, Microbiomes and their Role in Alzheimer's Disease. Front Aging Neurosci 2017; 9:336. [PMID: 29114218 PMCID: PMC5660720 DOI: 10.3389/fnagi.2017.00336] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/02/2017] [Indexed: 12/11/2022] Open
Abstract
As far back as the eighteenth and early nineteenth centuries, microbial infections were responsible for vast numbers of deaths. The trend reversed with the introduction of antibiotics coinciding with longer life. Increased life expectancy however, accompanied the emergence of age related chronic inflammatory states including the sporadic form of Alzheimer's disease (AD). Taken together, the true challenge of retaining health into later years of life now appears to lie in delaying and/or preventing the progression of chronic inflammatory diseases, through identifying and influencing modifiable risk factors. Diverse pathogens, including periodontal bacteria have been associated with AD brains. Amyloid-beta (Aβ) hallmark protein of AD may be a consequence of infection, called upon due to its antimicrobial properties. Up to this moment in time, a lack of understanding and knowledge of a microbiome associated with AD brain has ensured that the role pathogens may play in this neurodegenerative disease remains unresolved. The oral microbiome embraces a range of diverse bacterial phylotypes, which especially in vulnerable individuals, will excite and perpetuate a range of inflammatory conditions, to a wide range of extra-oral body tissues and organs specific to their developing pathophysiology, including the brain. This offers the tantalizing opportunity that by controlling the oral-specific microbiome; clinicians may treat or prevent a range of chronic inflammatory diseases orally. Evolution has equipped the human host to combat infection/disease by providing an immune system, but Porphyromonas gingivalis and selective spirochetes, have developed immune avoidance strategies threatening the host-microbe homeostasis. It is clear from longitudinal monitoring of patients that chronic periodontitis contributes to declining cognition. The aim here is to discuss the contribution from opportunistic pathogens of the periodontal microbiome, and highlight the challenges, the host faces, when dealing with unresolvable oral infections that may lead to clinical manifestations that are characteristic for AD.
Collapse
Affiliation(s)
- Anna B. Pritchard
- Dementia & Neurodegenerative Diseases Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, United Kingdom
| | - StJohn Crean
- Dementia & Neurodegenerative Diseases Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, United Kingdom
| | - Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Sim K. Singhrao
- Dementia & Neurodegenerative Diseases Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, United Kingdom
| |
Collapse
|
73
|
Liao S, De A, Thompson T, Chapman L, Bitoun JP, Yao X, Yu Q, Ma F, Wen ZT. Expression of BrpA in Streptococcus mutans is regulated by FNR-box mediated repression. Mol Oral Microbiol 2017; 32:517-525. [PMID: 28744965 DOI: 10.1111/omi.12193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2017] [Indexed: 11/30/2022]
Abstract
Our previous studies showed that brpA in Streptococcus mutans, which encodes a member of the LytR-CpsA-Psr family of proteins, can be co-transcribed with brpB upstream as a bicistronic operon, and the intergenic region also has strong promoter activity. To elucidate how brpA expression is regulated, the promoter regions were analyzed using polymerase chain reaction-based deletions and site-directed mutagenesis and a promoterless luciferase gene as a reporter. Allelic exchange mutagenesis was also used to examine genes encoding putative trans-acting factors, and the impact of such mutations on brpA expression was analyzed by reporter assays. Multiple elements in the short brpA promoter (nucleotide -1 to -344 relative to start cordon ATG) were shown to have a major impact on brpA expression, including an FNR-box, for a putative binding site of an FNR-type of transcriptional regulator. When compared with the intact brpA promoter, mutations of the highly conserved nucleotides in FNR-box from TTGATgtttAcCtt to TTACAgaaaGtTac resulted in 1362-fold increases of luciferase activity (P < .001), indicative of the FNR-box-mediated repression as a major mechanism in regulation of brpA expression. When luciferase reporter was fused to the upstream brpBA promoter (nucleotides -784 to -1144), luciferase activity was decreased by 4.5-fold (P < .001) in the brpA mutant, TW14D, and by 67.7-fold (P < .001) in the brpB mutant, JB409, compared with the wild-type, UA159. However, no such effects were observed when the reporter gene was fused to the short brpA promoter and its derivatives. These results also suggest that brpA expression in S. mutans is auto-regulated through the upstream brpBA promoter.
Collapse
Affiliation(s)
- S Liao
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - A De
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - T Thompson
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - L Chapman
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - J P Bitoun
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - X Yao
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Q Yu
- Department of Biostatistics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - F Ma
- Center for Virology, University of Nebraska, Lincoln, NE, USA
| | - Z T Wen
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
74
|
Yu J, Yang H, Li K, Ren H, Lei J, Huang C. Development of Epigallocatechin-3-gallate-Encapsulated Nanohydroxyapatite/Mesoporous Silica for Therapeutic Management of Dentin Surface. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25796-25807. [PMID: 28703572 DOI: 10.1021/acsami.7b06597] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In dental clinic, unsatisfactory management of the dentin surface after dentin exposure often leads to the occurrence of dentin hypersensitivity and caries. Current approaches can occlude the tubules on the dentin surface to relieve dentin hypersensitivity; however, the blocked tubules are generally weak in combating daily tooth erosion and abrasion. Moreover, cariogenic bacteria, such as Streptococcus mutans, produce biofilm on the dentin surface, causing caries and compromising the tubules' sealing efficacy. To overcome this problem, the present study focused on establishing a versatile biomaterial, epigallocatechin-3-gallate-encapsulated nanohydroxyapatite/mesoporous silica nanoparticle (EGCG@nHAp@MSN), for therapeutic management of the dentin surface. The effectiveness of the biomaterial on dentinal tubule occlusion, including resistances against acid and abrasion, was evaluated by field-emission scanning electron microscopy (FESEM) and dentin permeability measurement. The inhibitory capability of the biomaterial on S. mutans biofilm formation was investigated by confocal laser scanning microscopy (CLSM), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony forming units (CFU) counts, and FESEM. Results demonstrated for the first time that the use of EGCG@nHAp@MSN on the dentin surface was capable of effectively occluding dentinal tubules, reducing dentin permeability, and achieving favorable acid- and abrasion-resistant stability. Furthermore, EGCG@nHAp@MSN held the capability to continuously release EGCG, Ca, and P, and significantly inhibit the formation and growth of S. mutans biofilm on the dentin surface. Thus, the development of EGCG@nHAp@MSN bridges the gap between multifunctional concept and dental clinical practice and is promising in providing dentists a therapeutic strategy for the management of the dentin surface to counter dentin hypersensitivity and caries.
Collapse
Affiliation(s)
- Jian Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| | - Hongye Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| | - Kang Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| | - Hongyu Ren
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| | - Jinmei Lei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University , Wuhan 430079, China
| |
Collapse
|
75
|
Abstract
Curli are functional amyloids produced by proteobacteria like Escherichia coli as part of the extracellular matrix that holds cells together into biofilms. The molecular events that occur during curli nucleation and fiber extension remain largely unknown. Combining observations from curli amyloidogenesis in bulk solutions with real-time in situ nanoscopic imaging at the single-fiber level, we show that curli display polar growth, and we detect two kinetic regimes of fiber elongation. Single fibers exhibit stop-and-go dynamics characterized by bursts of steady-state growth alternated with periods of stagnation. At high subunit concentrations, fibers show constant, unperturbed burst growth. Curli follow a one-step nucleation process in which monomers contemporaneously fold and oligomerize into minimal fiber units that have growth characteristics identical to those of the mature fibrils. Kinetic data and interaction studies of curli fibrillation in the presence of the natural inhibitor CsgC show that the inhibitor binds curli fibers and predominantly acts at the level of fiber elongation.
Collapse
|
76
|
Besingi RN, Wenderska IB, Senadheera DB, Cvitkovitch DG, Long JR, Wen ZT, Brady LJ. Functional amyloids in Streptococcus mutans, their use as targets of biofilm inhibition and initial characterization of SMU_63c. MICROBIOLOGY-SGM 2017; 163:488-501. [PMID: 28141493 DOI: 10.1099/mic.0.000443] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Amyloids have been identified as functional components of the extracellular matrix of bacterial biofilms. Streptococcus mutans is an established aetiologic agent of dental caries and a biofilm dweller. In addition to the previously identified amyloidogenic adhesin P1 (also known as AgI/II, PAc), we show that the naturally occurring antigen A derivative of S. mutans wall-associated protein A (WapA) and the secreted protein SMU_63c can also form amyloid fibrils. P1, WapA and SMU_63c were found to significantly influence biofilm development and architecture, and all three proteins were shown by immunogold electron microscopy to reside within the fibrillar extracellular matrix of the biofilms. We also showed that SMU_63c functions as a negative regulator of biofilm cell density and genetic competence. In addition, the naturally occurring C-terminal cleavage product of P1, C123 (also known as AgII), was shown to represent the amyloidogenic moiety of this protein. Thus, P1 and WapA both represent sortase substrates that are processed to amyloidogenic truncation derivatives. Our current results suggest a novel mechanism by which certain cell surface adhesins are processed and contribute to the amyloidogenic capability of S. mutans. We further demonstrate that the polyphenolic small molecules tannic acid and epigallocatechin-3-gallate, and the benzoquinone derivative AA-861, which all inhibit amyloid fibrillization of C123 and antigen A in vitro, also inhibit S. mutans biofilm formation via P1- and WapA-dependent mechanisms, indicating that these proteins serve as therapeutic targets of anti-amyloid compounds.
Collapse
Affiliation(s)
- Richard N Besingi
- Department of Oral Biology, University of Florida, Gainesville, FL, USA
| | - Iwona B Wenderska
- Department of Oral Microbiology, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Dilani B Senadheera
- Department of Oral Microbiology, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Dennis G Cvitkovitch
- Department of Oral Microbiology, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Joanna R Long
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Zezhang T Wen
- Department of Comprehensive Dentistry and Biomaterials and Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - L Jeannine Brady
- Department of Oral Biology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
77
|
Ansari JM, Abraham NM, Massaro J, Murphy K, Smith-Carpenter J, Fikrig E. Anti-Biofilm Activity of a Self-Aggregating Peptide against Streptococcus mutans. Front Microbiol 2017; 8:488. [PMID: 28392782 PMCID: PMC5364132 DOI: 10.3389/fmicb.2017.00488] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/09/2017] [Indexed: 12/13/2022] Open
Abstract
Streptococcus mutans is the primary agent of dental cavities, in large part due to its ability to adhere to teeth and create a molecular scaffold of glucan polysaccharides on the tooth surface. Disrupting the architecture of S. mutans biofilms could help undermine the establishment of biofilm communities that cause cavities and tooth decay. Here we present a synthetic peptide P1, derived from a tick antifreeze protein, which significantly reduces S. mutans biofilm formation. Incubating cells with this peptide decreased biofilm biomass by approximately 75% in both a crystal violet microplate assay and an in vitro tooth model using saliva-coated hydroxyapatite discs. Bacteria treated with peptide P1 formed irregular biofilms with disconnected aggregates of cells and exopolymeric matrix that readily detached from surfaces. Peptide P1 can bind directly to S. mutans cells but does not possess bactericidal activity. Anti-biofilm activity was correlated with peptide aggregation and β-sheet formation in solution, and alternative synthetic peptides of different lengths or charge distribution did not inhibit biofilms. This anti-biofilm peptide interferes with S. mutans biofilm formation and architecture, and may have future applications in preventing bacterial buildup on teeth.
Collapse
Affiliation(s)
| | - Nabil M Abraham
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New HavenCT, USA; Howard Hughes Medical Institute, Chevy ChaseMD, USA
| | - Jenna Massaro
- Department of Biology, Fairfield University, Fairfield CT, USA
| | - Kelsey Murphy
- Department of Biology, Fairfield University, Fairfield CT, USA
| | | | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New HavenCT, USA; Howard Hughes Medical Institute, Chevy ChaseMD, USA
| |
Collapse
|
78
|
Markande AR, Nerurkar AS. Bioemulsifier (BE-AM1) produced by Solibacillus silvestris AM1 is a functional amyloid that modulates bacterial cell-surface properties. BIOFOULING 2016; 32:1153-1162. [PMID: 27669827 DOI: 10.1080/08927014.2016.1232716] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/29/2016] [Indexed: 06/06/2023]
Abstract
A novel estuarine bacterial strain, Solibacillus silvestris AM1, produces an extracellular, thermostable and fibrous, glycoprotein bioemulsifier (BE-AM1). The amyloid nature of the bioemulsifier (BE-AM1) was confirmed by biophysical techniques (Congo red based polarization microscopy, ThioflavinS based fluorescent microscopy, fibrous arrangement in transmission electron microscopy and secondary structure measurement by FTIR and CD spectrum analysis). Cell-bound BE-AM1 production by S. silvestris AM1 during the mid-logarithmic phase of growth coincided with a decrease in cell surface hydrophobicity, and an increase in cell autoaggregation and biofilm formation. It was observed that the total interfacial interaction energy ([Formula: see text]) for the surface of the bioemulsifier producing S. silvestris AM1 and different derivatized surfaces of polystyrene (silanized and sulfonated) was found to support biofilm formation. This study has revealed that the BE-AM1, a bacterial bioemulsifier, is a functional amyloid and has a role in biofilm formation and cell surface modulation in S. silvestris AM1.
Collapse
Affiliation(s)
- A R Markande
- a Department of Microbiology and Biotechnology Centre, Faculty of Science , The Maharaja Sayajirao University of Baroda , Vadodara , India
| | - A S Nerurkar
- a Department of Microbiology and Biotechnology Centre, Faculty of Science , The Maharaja Sayajirao University of Baroda , Vadodara , India
| |
Collapse
|
79
|
Pistollato F, Sumalla Cano S, Elio I, Masias Vergara M, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev 2016; 74:624-634. [PMID: 27634977 DOI: 10.1093/nutrit/nuw023] [Citation(s) in RCA: 358] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
It has been hypothesized that alterations in the composition of the gut microbiota might be associated with the onset of certain human pathologies, such as Alzheimer disease, a neurodegenerative syndrome associated with cerebral accumulation of amyloid-β fibrils. It has been shown that bacteria populating the gut microbiota can release significant amounts of amyloids and lipopolysaccharides, which might play a role in the modulation of signaling pathways and the production of proinflammatory cytokines related to the pathogenesis of Alzheimer disease. Additionally, nutrients have been shown to affect the composition of the gut microbiota as well as the formation and aggregation of cerebral amyloid-β. This suggests that modulating the gut microbiome and amyloidogenesis through specific nutritional interventions might prove to be an effective strategy to prevent or reduce the risk of Alzheimer disease. This review examines the possible role of the gut in the dissemination of amyloids, the role of the gut microbiota in the regulation of the gut-brain axis, the potential amyloidogenic properties of gut bacteria, and the possible impact of nutrients on modulation of microbiota composition and amyloid formation in relation to the pathogenesis of Alzheimer disease.
Collapse
Affiliation(s)
- Francesca Pistollato
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - Sandra Sumalla Cano
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - Iñaki Elio
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - Manuel Masias Vergara
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - Francesca Giampieri
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy.
| | - Maurizio Battino
- F. Pistollato, S.S. Cano, I. Elio, M.M. Vergara, F. Giampieri, and M. Battino are with the Centre for Nutrition and Health, Universidad Europea del Atlántico, Santander, Spain. S.S. Cano and I. Elio are with the Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico and the Fundacion Universitaria Iberoamericana (FUNIBER), Barcelona, Spain. M.M. Vergara is with the Universidad Internacional Iberoamericana (UNINI), Arecibo, Puerto Rico, USA. F. Giampieri and M. Battino are with the Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
80
|
Abstract
Recent insights into bacterial biofilm matrix structures have induced a paradigm shift toward the recognition of amyloid fibers as common building block structures that confer stability to the exopolysaccharide matrix. Here we describe the functional amyloid systems related to biofilm matrix formation in both Gram-negative and Gram-positive bacteria and recent knowledge regarding the interaction of amyloids with other biofilm matrix components such as extracellular DNA (eDNA) and the host immune system. In addition, we summarize the efforts to identify compounds that target amyloid fibers for therapeutic purposes and recent developments that take advantage of the amyloid structure to engineer amyloid fibers of bacterial biofilm matrices for biotechnological applications.
Collapse
|
81
|
Althani AA, Marei HE, Hamdi WS, Nasrallah GK, El Zowalaty ME, Al Khodor S, Al-Asmakh M, Abdel-Aziz H, Cenciarelli C. Human Microbiome and its Association With Health and Diseases. J Cell Physiol 2016; 231:1688-1694. [PMID: 26660761 DOI: 10.1002/jcp.25284] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/09/2015] [Indexed: 12/29/2022]
Abstract
Human microbiota are distinct communities of microorganisms that resides at different body niches. Exploration of the human microbiome has become a reality due to the availability of powerful metagenomics and metatranscriptomic analysis technologies. Recent advances in sequencing and bioinformatics over the past decade help provide a deep insight into the nature of the host-microbial interactions and identification of potential deriver genes and pathways associated with human health, well-being, and predisposition to different diseases. In the present review, we outline recent studies devoted to elucidate the possible link between the microbiota and various type of diseases. The present review also highlights the potential utilization of microbiota as a potential therapeutic option to treat a wide array of human diseases. J. Cell. Physiol. 231: 1688-1694, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Asmaa A Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Hany E Marei
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Wedad S Hamdi
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | | | - Souhaila Al Khodor
- Infectious Disease Unit, Division of Translational Medicine, SIDRA Medical and Research Center, Qatar
| | - Maha Al-Asmakh
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Hassan Abdel-Aziz
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Carlo Cenciarelli
- CNR-Institute of Translational Pharmacology, Via Fosso del Cavaliere, Roma, Italy
| |
Collapse
|
82
|
Staphylococcal Bap Proteins Build Amyloid Scaffold Biofilm Matrices in Response to Environmental Signals. PLoS Pathog 2016; 12:e1005711. [PMID: 27327765 PMCID: PMC4915627 DOI: 10.1371/journal.ppat.1005711] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
Biofilms are communities of bacteria that grow encased in an extracellular matrix that often contains proteins. The spatial organization and the molecular interactions between matrix scaffold proteins remain in most cases largely unknown. Here, we report that Bap protein of Staphylococcus aureus self-assembles into functional amyloid aggregates to build the biofilm matrix in response to environmental conditions. Specifically, Bap is processed and fragments containing at least the N-terminus of the protein become aggregation-prone and self-assemble into amyloid-like structures under acidic pHs and low concentrations of calcium. The molten globule-like state of Bap fragments is stabilized upon binding of the cation, hindering its self-assembly into amyloid fibers. These findings define a dual function for Bap, first as a sensor and then as a scaffold protein to promote biofilm development under specific environmental conditions. Since the pH-driven multicellular behavior mediated by Bap occurs in coagulase-negative staphylococci and many other bacteria exploit Bap-like proteins to build a biofilm matrix, the mechanism of amyloid-like aggregation described here may be widespread among pathogenic bacteria. Major components of the biofilm matrix scaffold are proteins that assemble to create a unified structure that maintain bacteria attached to each other and to surfaces. We provide evidence that a surface protein present in several staphylococcal species forms functional amyloid aggregates to build the biofilm matrix in response to specific environmental conditions. Under low Ca2+ concentrations and acidic pH, Bap is processed and forms insoluble aggregates with amyloidogenic properties. When the Ca2+ concentration increases, metal-coordinated Bap adopts a structurally more stable conformation and as a consequence, the N-terminal region is unable to assemble into amyloid aggregates. The control of Bap cleavage and assembly helps to regulate biofilm matrix development as a function of environmental changes.
Collapse
|
83
|
Tang W, Bhatt A, Smith AN, Crowley PJ, Brady LJ, Long JR. Specific binding of a naturally occurring amyloidogenic fragment of Streptococcus mutans adhesin P1 to intact P1 on the cell surface characterized by solid state NMR spectroscopy. JOURNAL OF BIOMOLECULAR NMR 2016; 64:153-64. [PMID: 26837620 PMCID: PMC4756430 DOI: 10.1007/s10858-016-0017-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/24/2016] [Indexed: 06/05/2023]
Abstract
The P1 adhesin (aka Antigen I/II or PAc) of the cariogenic bacterium Streptococcus mutans is a cell surface-localized protein involved in sucrose-independent adhesion and colonization of the tooth surface. The immunoreactive and adhesive properties of S. mutans suggest an unusual functional quaternary ultrastructure comprised of intact P1 covalently attached to the cell wall and interacting with non-covalently associated proteolytic fragments thereof, particularly the ~57-kDa C-terminal fragment C123 previously identified as Antigen II. S. mutans is capable of amyloid formation when grown in a biofilm and P1 is among its amyloidogenic proteins. The C123 fragment of P1 readily forms amyloid fibers in vitro suggesting it may play a role in the formation of functional amyloid during biofilm development. Using wild-type and P1-deficient strains of S. mutans, we demonstrate that solid state NMR (ssNMR) spectroscopy can be used to (1) globally characterize cell walls isolated from a Gram-positive bacterium and (2) characterize the specific binding of heterologously expressed, isotopically-enriched C123 to cell wall-anchored P1. Our results lay the groundwork for future high-resolution characterization of the C123/P1 ultrastructure and subsequent steps in biofilm formation via ssNMR spectroscopy, and they support an emerging model of S. mutans colonization whereby quaternary P1-C123 interactions confer adhesive properties important to binding to immobilized human salivary agglutinin.
Collapse
Affiliation(s)
- Wenxing Tang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Avni Bhatt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Adam N Smith
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Paula J Crowley
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - L Jeannine Brady
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA.
| | - Joanna R Long
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
84
|
Schwartz K, Ganesan M, Payne DE, Solomon MJ, Boles BR. Extracellular DNA facilitates the formation of functional amyloids in Staphylococcus aureus biofilms. Mol Microbiol 2015; 99:123-34. [PMID: 26365835 DOI: 10.1111/mmi.13219] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2015] [Indexed: 11/27/2022]
Abstract
Persistent staphylococcal infections often involve surface-associated communities called biofilms. Staphylococcus aureus biofilm development is mediated by the co-ordinated production of the biofilm matrix, which can be composed of polysaccharides, extracellular DNA (eDNA) and proteins including amyloid fibers. The nature of the interactions between matrix components, and how these interactions contribute to the formation of matrix, remain unclear. Here we show that the presence of eDNA in S. aureus biofilms promotes the formation of amyloid fibers. Conditions or mutants that do not generate eDNA result in lack of amyloids during biofilm growth despite the amyloidogeneic subunits, phenol soluble modulin peptides, being produced. In vitro studies revealed that the presence of DNA promotes amyloid formation by PSM peptides. Thus, this work exposes a previously unacknowledged interaction between biofilm matrix components that furthers our understanding of functional amyloid formation and S. aureus biofilm biology.
Collapse
Affiliation(s)
- Kelly Schwartz
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Mahesh Ganesan
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - David E Payne
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael J Solomon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Blaise R Boles
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
85
|
Chan CXJ, Joseph IG, Huang A, Jackson DN, Lipke PN. Quantitative Analyses of Force-Induced Amyloid Formation in Candida albicans Als5p: Activation by Standard Laboratory Procedures. PLoS One 2015; 10:e0129152. [PMID: 26047318 PMCID: PMC4457901 DOI: 10.1371/journal.pone.0129152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/05/2015] [Indexed: 11/18/2022] Open
Abstract
Candida albicans adhesins have amyloid-forming sequences. In Als5p, these amyloid sequences cluster cell surface adhesins to create high avidity surface adhesion nanodomains. Such nanodomains form after force is applied to the cell surface by atomic force microscopy or laminar flow. Here we report centrifuging and resuspending S. cerevisiae cells expressing Als5p led to 1.7-fold increase in initial rate of adhesion to ligand coated beads. Furthermore, mechanical stress from vortex-mixing of Als5p cells or C. albicans cells also induced additional formation of amyloid nanodomains and consequent activation of adhesion. Vortex-mixing for 60 seconds increased the initial rate of adhesion 1.6-fold. The effects of vortex-mixing were replicated in heat-killed cells as well. Activation was accompanied by increases in thioflavin T cell surface fluorescence measured by flow cytometry or by confocal microscopy. There was no adhesion activation in cells expressing amyloid-impaired Als5pV326N or in cells incubated with inhibitory concentrations of anti-amyloid dyes. Together these results demonstrated the activation of cell surface amyloid nanodomains in yeast expressing Als adhesins, and further delineate the forces that can activate adhesion in vivo. Consequently there is quantitative support for the hypothesis that amyloid forming adhesins act as both force sensors and effectors.
Collapse
Affiliation(s)
- Cho X. J. Chan
- Biology Department, Brooklyn College City University of New York, New York, New York, United States of America
- The Graduate Center, City University of New York, New York, New York, United States of America
- Haskins Laboratories and the Department of Chemistry and Physical Sciences, Pace University, New York, New York, United States of America
| | - Ivor G. Joseph
- Biology Department, Brooklyn College City University of New York, New York, New York, United States of America
| | - Andy Huang
- Biology Department, Brooklyn College City University of New York, New York, New York, United States of America
| | - Desmond N. Jackson
- Biology Department, Brooklyn College City University of New York, New York, New York, United States of America
| | - Peter N. Lipke
- Biology Department, Brooklyn College City University of New York, New York, New York, United States of America
- The Graduate Center, City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
86
|
Morphological and proteomic analyses of the biofilms generated by Streptococcus mutans isolated from caries-active and caries-free adults. J Dent Sci 2015. [DOI: 10.1016/j.jds.2014.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
87
|
Ramsugit S, Pillay M. Pili of Mycobacterium tuberculosis: current knowledge and future prospects. Arch Microbiol 2015; 197:737-44. [PMID: 25975850 DOI: 10.1007/s00203-015-1117-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 11/29/2022]
Abstract
Many pathogenic bacteria express filamentous appendages, termed pili, on their surface. These organelles function in several important bacterial processes, including mediating bacterial interaction with, and colonization of the host, signalling events, locomotion, DNA uptake, electric conductance, and biofilm formation. In the last decade, it has been established that the tuberculosis-causing bacterium, Mycobacterium tuberculosis, produces two pili types: curli and type IV pili. In this paper, we review studies on M. tuberculosis pili, highlighting their structure and biological significance to M. tuberculosis pathogenesis, and discuss their potential as targets for therapeutic intervention and diagnostic test development.
Collapse
Affiliation(s)
- Saiyur Ramsugit
- Medical Microbiology and Infection Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st Floor Doris Duke Medical Research Institute, Private Bag 7, Congella, Durban, 4013, South Africa
| | | |
Collapse
|
88
|
Abstract
Amyloids are insoluble fibrillar protein deposits with an underlying cross-β structure initially discovered in the context of human diseases. However, it is now clear that the same fibrillar structure is used by many organisms, from bacteria to humans, in order to achieve a diverse range of biological functions. These functions include structure and protection (e.g. curli and chorion proteins, and insect and spider silk proteins), aiding interface transitions and cell-cell recognition (e.g. chaplins, rodlins and hydrophobins), protein control and storage (e.g. Microcin E492, modulins and PMEL), and epigenetic inheritance and memory [e.g. Sup35, Ure2p, HET-s and CPEB (cytoplasmic polyadenylation element-binding protein)]. As more examples of functional amyloid come to light, the list of roles associated with functional amyloids has continued to expand. More recently, amyloids have also been implicated in signal transduction [e.g. RIP1/RIP3 (receptor-interacting protein)] and perhaps in host defence [e.g. aDrs (anionic dermaseptin) peptide]. The present chapter discusses in detail functional amyloids that are used in Nature by micro-organisms, non-mammalian animals and mammals, including the biological roles that they play, their molecular composition and how they assemble, as well as the coping strategies that organisms have evolved to avoid the potential toxicity of functional amyloid.
Collapse
|
89
|
Hobley L, Harkins C, MacPhee CE, Stanley-Wall NR. Giving structure to the biofilm matrix: an overview of individual strategies and emerging common themes. FEMS Microbiol Rev 2015; 39:649-69. [PMID: 25907113 PMCID: PMC4551309 DOI: 10.1093/femsre/fuv015] [Citation(s) in RCA: 361] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2015] [Indexed: 01/24/2023] Open
Abstract
Biofilms are communities of microbial cells that underpin diverse processes including sewage bioremediation, plant growth promotion, chronic infections and industrial biofouling. The cells resident in the biofilm are encased within a self-produced exopolymeric matrix that commonly comprises lipids, proteins that frequently exhibit amyloid-like properties, eDNA and exopolysaccharides. This matrix fulfils a variety of functions for the community, from providing structural rigidity and protection from the external environment to controlling gene regulation and nutrient adsorption. Critical to the development of novel strategies to control biofilm infections, or the capability to capitalize on the power of biofilm formation for industrial and biotechnological uses, is an in-depth knowledge of the biofilm matrix. This is with respect to the structure of the individual components, the nature of the interactions between the molecules and the three-dimensional spatial organization. We highlight recent advances in the understanding of the structural and functional role that carbohydrates and proteins play within the biofilm matrix to provide three-dimensional architectural integrity and functionality to the biofilm community. We highlight, where relevant, experimental techniques that are allowing the boundaries of our understanding of the biofilm matrix to be extended using Escherichia coli, Staphylococcus aureus, Vibrio cholerae, and Bacillus subtilis as exemplars. Examining the structure and function of the biofilm extracellular matrix.
Collapse
Affiliation(s)
- Laura Hobley
- Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Catriona Harkins
- Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Cait E MacPhee
- James Clerk Maxwell Building, School of Physics, University of Edinburgh, Edinburgh EH9 3JZ, UK
| | - Nicola R Stanley-Wall
- Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
90
|
Wen ZT, Bitoun JP, Liao S. PBP1a-deficiency causes major defects in cell division, growth and biofilm formation by Streptococcus mutans. PLoS One 2015; 10:e0124319. [PMID: 25880908 PMCID: PMC4399832 DOI: 10.1371/journal.pone.0124319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/27/2015] [Indexed: 01/26/2023] Open
Abstract
Streptococcus mutans, a key etiological agent of human dental caries, lives almost exclusively on the tooth surface in plaque biofilms and is known for its ability to survive and respond to various environmental insults, including low pH, and antimicrobial agents from other microbes and oral care products. In this study, a penicillin-binding protein (PBP1a)-deficient mutant, strain JB467, was generated by allelic replacement mutagenesis and analyzed for the effects of such a deficiency on S. mutans’ stress tolerance response and biofilm formation. Our results so far have shown that PBP1a-deficiency in S. mutans affects growth of the deficient mutant, especially at acidic and alkaline pHs. As compared to the wild-type, UA159, the PBP1a-deficient mutant, JB467, had a reduced growth rate at pH 6.2 and did not grow at all at pH 8.2. Unlike the wild-type, the inclusion of paraquat in growth medium, especially at 2 mM or above, significantly reduced the growth rate of the mutant. Acid killing assays showed that the mutant was 15-fold more sensitive to pH 2.8 than the wild-type after 30 minutes. In a hydrogen peroxide killing assay, the mutant was 16-fold more susceptible to hydrogen peroxide (0.2%, w/v) after 90 minutes than the wild-type. Relative to the wild-type, the mutant also had an aberrant autolysis rate, indicative of compromises in cell envelope integrity. As analyzed using on 96-well plate model and spectrophotometry, biofilm formation by the mutant was decreased significantly, as compared to the wild-type. Consistently, Field Emission-SEM analysis also showed that the PBP1a-deficient mutant had limited capacity to form biofilms. TEM analysis showed that PBP1a mutant existed primarily in long rod-like cells and cells with multiple septa, as compared to the coccal wild-type. The results presented here highlight the importance of pbp1a in cell morphology, stress tolerance, and biofilm formation in S. mutans.
Collapse
Affiliation(s)
- Zezhang T. Wen
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
- * E-mail:
| | - Jacob P. Bitoun
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
| | - Sumei Liao
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States of America
| |
Collapse
|
91
|
Heim KP, Sullan RMA, Crowley PJ, El-Kirat-Chatel S, Beaussart A, Tang W, Besingi R, Dufrene YF, Brady LJ. Identification of a supramolecular functional architecture of Streptococcus mutans adhesin P1 on the bacterial cell surface. J Biol Chem 2015; 290:9002-19. [PMID: 25666624 DOI: 10.1074/jbc.m114.626663] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Indexed: 12/29/2022] Open
Abstract
P1 (antigen I/II) is a sucrose-independent adhesin of Streptococcus mutans whose functional architecture on the cell surface is not fully understood. S. mutans cells subjected to mechanical extraction were significantly diminished in adherence to immobilized salivary agglutinin but remained immunoreactive and were readily aggregated by fluid-phase salivary agglutinin. Bacterial adherence was restored by incubation of postextracted cells with P1 fragments that contain each of the two known adhesive domains. In contrast to untreated cells, glutaraldehyde-treated bacteria gained reactivity with anti-C-terminal monoclonal antibodies (mAbs), whereas epitopes recognized by mAbs against other portions of the molecule were masked. Surface plasmon resonance experiments demonstrated the ability of apical and C-terminal fragments of P1 to interact. Binding of several different anti-P1 mAbs to unfixed cells triggered release of a C-terminal fragment from the bacterial surface, suggesting a novel mechanism of action of certain adherence-inhibiting antibodies. We also used atomic force microscopy-based single molecule force spectroscopy with tips bearing various mAbs to elucidate the spatial organization and orientation of P1 on living bacteria. The similar rupture lengths detected using mAbs against the head and C-terminal regions, which are widely separated in the tertiary structure, suggest a higher order architecture in which these domains are in close proximity on the cell surface. Taken together, our results suggest a supramolecular organization in which additional P1 polypeptides, including the C-terminal segment originally identified as antigen II, associate with covalently attached P1 to form the functional adhesive layer.
Collapse
Affiliation(s)
- Kyle P Heim
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Ruby May A Sullan
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Paula J Crowley
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Sofiane El-Kirat-Chatel
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Audrey Beaussart
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Wenxing Tang
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Richard Besingi
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Yves F Dufrene
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - L Jeannine Brady
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| |
Collapse
|
92
|
Bhatty M, Cruz MR, Frank KL, Gomez JAL, Andrade F, Garsin DA, Dunny GM, Kaplan HB, Christie PJ. Enterococcus faecalis pCF10-encoded surface proteins PrgA, PrgB (aggregation substance) and PrgC contribute to plasmid transfer, biofilm formation and virulence. Mol Microbiol 2014; 95:660-77. [PMID: 25431047 DOI: 10.1111/mmi.12893] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2014] [Indexed: 01/24/2023]
Abstract
Enterococcus faecalis pCF10 transfers at high frequencies upon pheromone induction of the prgQ transfer operon. This operon codes for three cell wall-anchored proteins - PrgA, PrgB (aggregation substance) and PrgC - and a type IV secretion system through which the plasmid is delivered to recipient cells. Here, we defined the contributions of the Prg surface proteins to plasmid transfer, biofilm formation and virulence using the Caenorhabditis elegans infection model. We report that a combination of PrgB and extracellular DNA (eDNA), but not PrgA or PrgC, was required for extensive cellular aggregation and pCF10 transfer at wild-type frequencies. In addition to PrgB and eDNA, production of PrgA was necessary for extensive binding of enterococci to abiotic surfaces and development of robust biofilms. However, although PrgB is a known virulence factor in mammalian infection models, we determined that PrgA and PrgC, but not PrgB, were required for efficient killing in the worm infection model. We propose that the pheromone-responsive, conjugative plasmids of E. faecalis have retained Prg-like surface functions over evolutionary time for attachment, colonization and robust biofilm development. In natural settings, these biofilms are polymicrobial in composition and constitute optimal environments for signal exchange, mating pair formation and widespread lateral gene transfer.
Collapse
Affiliation(s)
- Minny Bhatty
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Liebert A, Bicknell B, Adams R. Prion Protein Signaling in the Nervous System—A Review and Perspective. ACTA ACUST UNITED AC 2014. [DOI: 10.4137/sti.s12319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Prion protein (PrPC) was originally known as the causative agent of transmissible spongiform encephalopathy (TSE) but with recent research, its true function in cells is becoming clearer. It is known to act as a scaffolding protein, binding multiple ligands at the cell membrane and to be involved in signal transduction, passing information from the extracellular matrix (ECM) to the cytoplasm. Its role in the coordination of transmitters at the synapse, glyapse, and gap junction and in short- and long-range neurotrophic signaling gives PrPC a major part in neural transmission and nervous system signaling. It acts to regulate cellular function in multiple targets through its role as a controller of redox status and calcium ion flux. Given the importance of PrPC in cell physiology, this review considers its potential role in disease apart from TSE. The putative functions of PrPC point to involvement in neurodegenerative disease, neuropathic pain, chronic headache, and inflammatory disease including neuroinflammatory disease of the nervous system. Potential targets for the treatment of disease influenced by PrPC are discussed.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Health Science, University of Sydney, Australia
| | - Brian Bicknell
- Faculty of Health Science, Australian Catholic University, Australia
| | | |
Collapse
|
94
|
Syed AK, Boles BR. Fold modulating function: bacterial toxins to functional amyloids. Front Microbiol 2014; 5:401. [PMID: 25136340 PMCID: PMC4118032 DOI: 10.3389/fmicb.2014.00401] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/16/2014] [Indexed: 12/11/2022] Open
Abstract
Many bacteria produce cytolytic toxins that target host cells or other competing microbes. It is well known that environmental factors control toxin expression, however, recent work suggests that some bacteria manipulate the fold of these protein toxins to control their function. The β-sheet rich amyloid fold is a highly stable ordered aggregate that many toxins form in response to specific environmental conditions. When in the amyloid state, toxins become inert, losing the cytolytic activity they display in the soluble form. Emerging evidence suggest that some amyloids function as toxin storage systems until they are again needed, while other bacteria utilize amyloids as a structural matrix component of biofilms. This amyloid matrix component facilitates resistance to biofilm disruptive challenges. The bacterial amyloids discussed in this review reveal an elegant system where changes in protein fold and solubility dictate the function of proteins in response to the environment.
Collapse
Affiliation(s)
- Adnan K Syed
- Department of Molecular Cellular and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| | - Blaise R Boles
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa Iowa City, IA, USA
| |
Collapse
|
95
|
Mishra S, Routray S, Kumar Sahu S, Bhusan Nanda S, Charan Sahu K. The role and efficacy of herbal antimicrobial agents in orthodontic treatment. J Clin Diagn Res 2014; 8:ZC12-4. [PMID: 25121056 PMCID: PMC4129259 DOI: 10.7860/jcdr/2014/7349.4464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 01/27/2014] [Indexed: 11/24/2022]
Abstract
AIMS To assess the effect of herbal antimicrobial agents on Streptococcus mutans count in biofilm formations during orthodontic treatment. MATERIALS AND METHODS We calculated the growth inhibition of oral bacteria in the orthodontic appliances after herbal antibacterial agents were placed in culture media. The Minimum Inhibitory Concentrations (MICs) of these agents on Streptococcus mutans growth were determined. After cultivating colonies of Streptococci in biofilm medium with these herbal antimicrobial agents and orthodontic attachments, viable cell counting was performed from the bacteria which were attached on them. Scanning electron microscopy (SEM) analysis of morphology was observed on bacterial cells which were attached to orthodontic attachments. The effects of these agents were then evaluated and recommendations were forwarded. RESULTS There was an increase in count of Streptococcus mutans with respect to the herbal antibacterial agents. CONCLUSION Despite the antibacterial functions of these herbal agents, there was increase in the biofilm formation caused by Streptococcus mutans to orthodontic bands, which had occurred most likely through upregulation of glucosyl transferase expression. These extracts may thus play an important role in increased bacterial attachment to orthodontic wires. Thus, this study was corroborative of an amalgamation of Ayurvedic therapy and Orthodontic treatment.
Collapse
Affiliation(s)
- Sumita Mishra
- Senior Lecturer, Department of Orthodontics, Institute of Dental Sciences, SOA University,Bhubaneswar, Odisha, India
| | - Samapika Routray
- Senior Lecturer, Department of Oral Pathology and Microbiology, Institute of Dental Sciences, SOA University, Bhubaneswar, Odisha, India
| | - Sanjeeb Kumar Sahu
- Professor, Department of Orthodontics, Institute of Dental Sciences, SOA University, Bhubaneswar, Odisha, India
| | - Smruti Bhusan Nanda
- Reader, Department of Orthodontics, Institute of Dental Sciences, SOA University, Bhubaneswar, Odisha, India
| | - Kanhu Charan Sahu
- Senior Lecturer, Department of Orthodontics, Institute of Dental Sciences, SOA University, Bhubaneswar, Odisha, India
| |
Collapse
|
96
|
Abstract
The conformational diseases, linked to protein aggregation into amyloid conformations, range from non-infectious neurodegenerative disorders, such as Alzheimer disease (AD), to highly infectious ones, such as human transmissible spongiform encephalopathies (TSEs). They are commonly known as prion diseases. However, since all amyloids could be considered prions (from those involved in cell-to-cell transmission to those responsible for real neuronal invasion), it is necessary to find an underlying cause of the different capacity to infect that each of the proteins prone to form amyloids has. As proposed here, both the intrinsic cytotoxicity and the number of nuclei of aggregation per cell could be key factors in this transmission capacity of each amyloid.
Collapse
Affiliation(s)
- Raimon Sabate
- Conformational Diseases Group; Department of Physical Chemistry; Faculty of Pharmacy; University of Barcelona (UB); Barcelona, Spain; Institut of Nanoscience and Nanotechnology of the University of Barcelona (IN2UB); Barcelona, Spain
| |
Collapse
|
97
|
|
98
|
Streptococcus mutans extracellular DNA is upregulated during growth in biofilms, actively released via membrane vesicles, and influenced by components of the protein secretion machinery. J Bacteriol 2014; 196:2355-66. [PMID: 24748612 DOI: 10.1128/jb.01493-14] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Streptococcus mutans, a major etiological agent of human dental caries, lives primarily on the tooth surface in biofilms. Limited information is available concerning the extracellular DNA (eDNA) as a scaffolding matrix in S. mutans biofilms. This study demonstrates that S. mutans produces eDNA by multiple avenues, including lysis-independent membrane vesicles. Unlike eDNAs from cell lysis that were abundant and mainly concentrated around broken cells or cell debris with floating open ends, eDNAs produced via the lysis-independent pathway appeared scattered but in a structured network under scanning electron microscopy. Compared to eDNA production of planktonic cultures, eDNA production in 5- and 24-h biofilms was increased by >3- and >1.6-fold, respectively. The addition of DNase I to growth medium significantly reduced biofilm formation. In an in vitro adherence assay, added chromosomal DNA alone had a limited effect on S. mutans adherence to saliva-coated hydroxylapatite beads, but in conjunction with glucans synthesized using purified glucosyltransferase B, the adherence was significantly enhanced. Deletion of sortase A, the transpeptidase that covalently couples multiple surface-associated proteins to the cell wall peptidoglycan, significantly reduced eDNA in both planktonic and biofilm cultures. Sortase A deficiency did not have a significant effect on membrane vesicle production; however, the protein profile of the mutant membrane vesicles was significantly altered, including reduction of adhesin P1 and glucan-binding proteins B and C. Relative to the wild type, deficiency of protein secretion and membrane protein insertion machinery components, including Ffh, YidC1, and YidC2, also caused significant reductions in eDNA.
Collapse
|
99
|
Abstract
We tell of a journey that led to discovery of amyloids formed by yeast cell adhesins and their importance in biofilms and host immunity. We begin with the identification of the adhesin functional amyloid-forming sequences that mediate fiber formation in vitro. Atomic force microscopy and confocal microscopy show 2-dimensional amyloid "nanodomains" on the surface of cells that are activated for adhesion. These nanodomains are arrays of adhesin molecules that bind multivalent ligands with high avidity. Nanodomains form when adhesin molecules are stretched in the AFM or under laminar flow. Treatment with antiamyloid perturbants or mutation of the amyloid sequence prevents adhesion nanodomain formation and activation. We are now discovering biological consequences. Adhesin nanodomains promote formation and maintenance of biofilms, which are microbial communities. Also, in abscesses within candidiasis patients, we find adhesin amyloids on the surface of the fungi. In both human infection and a Caenorhabditis elegans infection model, the presence of fungal surface amyloids elicits anti-inflammatory responses. Thus, this is a story of how fungal adhesins respond to extension forces through formation of cell surface amyloid nanodomains, with key consequences for biofilm formation and host responses.
Collapse
|
100
|
Peptide detection of fungal functional amyloids in infected tissue. PLoS One 2014; 9:e86067. [PMID: 24465872 PMCID: PMC3897640 DOI: 10.1371/journal.pone.0086067] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/09/2013] [Indexed: 01/09/2023] Open
Abstract
Many fungal cell adhesion proteins form functional amyloid patches on the surface of adhering cells. The Candida albicansAgglutinin-like sequence (Als) adhesins are exemplars for this phenomenon, and have amyloid forming sequences that are conserved between family members. The Als5p amyloid sequence mediates amyloid fibril formation and is critical for cell adhesion and biofilm formation, and is also present in the related adhesins Als1p and Als3p. We have developed a fluorescent peptide probe containing the conserved Als amyloid-forming sequence. This peptide bound specifically to yeast expressing Als5p, but not to cells lacking the adhesin. The probe bound to both yeast and hyphal forms of C. albicans. Δals1/Δals3 single and double deletion strains exhibited reduced fluorescence, indicating that probe binding required expression of these proteins. Additionally, the Als peptide specifically stained fungal cells in abscesses in autopsy sections. Counterstaining with calcofluor white showed colocalization with the amyloid peptide. In addition, fungi in autopsy sections derived from the gastrointestinal tract showed colocalization of the amyloid-specific dye thioflavin T and the fluorescent peptide. Collectively, our data demonstrate that we can exploit amyloid sequence specificity for detection of functional amyloids in situ.
Collapse
|