Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 184(23): 5740-5758.e17, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34735796

RESUMEN

Biofilms are community architectures adopted by bacteria inclusive of a self-formed extracellular matrix that protects resident bacteria from diverse environmental stresses and, in many species, incorporates extracellular DNA (eDNA) and DNABII proteins for structural integrity throughout biofilm development. Here, we present evidence that this eDNA-based architecture relies on the rare Z-form. Z-form DNA accumulates as biofilms mature and, through stabilization by the DNABII proteins, confers structural integrity to the biofilm matrix. Indeed, substances known to drive B-DNA into Z-DNA promoted biofilm formation whereas those that drive Z-DNA into B-DNA disrupted extant biofilms. Importantly, we demonstrated that the universal bacterial DNABII family of proteins stabilizes both bacterial- and host-eDNA in the Z-form in situ. A model is proposed that incorporates the role of Z-DNA in biofilm pathogenesis, innate immune response, and immune evasion.


Asunto(s)
Bacterias/genética , Biopelículas , ADN Bacteriano/química , Matriz Extracelular/metabolismo , Espacio Extracelular/química , Animales , Especificidad de Anticuerpos , Proteínas Bacterianas/metabolismo , Línea Celular , Chinchilla , ADN Cruciforme , Desoxirribonucleasas/metabolismo , Trampas Extracelulares/metabolismo , Humanos , Acetato de Tetradecanoilforbol/farmacología
2.
Antimicrob Agents Chemother ; 66(3): e0187721, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35007137

RESUMEN

New strategies to treat diseases in which biofilms contribute significantly to pathogenesis are needed, as biofilm-resident bacteria are highly recalcitrant to antibiotics due to physical biofilm architecture and a canonically quiescent metabolism, among many additional attributes. We, and others, have shown that when biofilms are dispersed or disrupted, bacteria released from biofilm residence are in a distinct physiologic state that, in part, renders these bacteria highly sensitive to killing by specific antibiotics. We sought to demonstrate the breadth of the ability of a recently humanized monoclonal antibody against an essential biofilm structural element (DNABII protein) to disrupt biofilms formed by respiratory tract pathogens and potentiate antibiotic-mediated killing of bacteria released from biofilm residence. Biofilms formed by six respiratory tract pathogens were significantly disrupted by the humanized monoclonal antibody in a dose- and time-dependent manner, as corroborated by confocal laser scanning microscopy (CLSM) imaging. Bacteria newly released from the biofilms of 3 of 6 species were significantly more sensitive than their planktonic counterparts to killing by 2 of 3 antibiotics currently used clinically and were now also equally as sensitive to killing by the 3rd antibiotic. The remaining 3 pathogens were significantly more susceptible to killing by all 3 antibiotics. A humanized monoclonal antibody directed against protective epitopes of a DNABII protein effectively released six diverse respiratory tract pathogens from biofilm residence in a phenotypic state that was now as, or significantly more, sensitive to killing by three antibiotics currently indicated for use clinically. These data support this targeted, combinatorial, species-agnostic therapy to mitigate chronic bacterial diseases.


Asunto(s)
Antibacterianos , Infecciones Bacterianas , Antibacterianos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Infecciones Bacterianas/microbiología , Biopelículas , Humanos , Sistema Respiratorio
3.
Proc Natl Acad Sci U S A ; 116(50): 25068-25077, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31767757

RESUMEN

Extracellular DNA (eDNA) is a critical component of the extracellular matrix of bacterial biofilms that protects the resident bacteria from environmental hazards, which includes imparting significantly greater resistance to antibiotics and host immune effectors. eDNA is organized into a lattice-like structure, stabilized by the DNABII family of proteins, known to have high affinity and specificity for Holliday junctions (HJs). Accordingly, we demonstrated that the branched eDNA structures present within the biofilms formed by NTHI in the middle ear of the chinchilla in an experimental otitis media model, and in sputum samples recovered from cystic fibrosis patients that contain multiple mixed bacterial species, possess an HJ-like configuration. Next, we showed that the prototypic Escherichia coli HJ-specific DNA-binding protein RuvA could be functionally exchanged for DNABII proteins in the stabilization of biofilms formed by 3 diverse human pathogens, uropathogenic E. coli, nontypeable Haemophilus influenzae, and Staphylococcus epidermidis Importantly, while replacement of DNABII proteins within the NTHI biofilm matrix with RuvA was shown to retain similar mechanical properties when compared to the control NTHI biofilm structure, we also demonstrated that biofilm eDNA matrices stabilized by RuvA could be subsequently undermined upon addition of the HJ resolvase complex, RuvABC, which resulted in significant biofilm disruption. Collectively, our data suggested that nature has recapitulated a functional equivalent of the HJ recombination intermediate to maintain the structural integrity of bacterial biofilms.


Asunto(s)
Biopelículas , ADN Cruciforme , Matriz Extracelular , Resolvasas de Unión Holliday , Recombinación Genética , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Chinchilla , ADN Helicasas , ADN Cruciforme/química , ADN Cruciforme/metabolismo , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Proteínas de Escherichia coli , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Resolvasas de Unión Holliday/química , Resolvasas de Unión Holliday/metabolismo , Otitis Media
4.
J Bacteriol ; 200(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29437850

RESUMEN

The oral cavity is home to a wide variety of bacterial species, both commensal, such as various streptococcal species, and pathogenic, such as Porphyromonas gingivalis, one of the main etiological agents of periodontal disease. Our understanding of how these bacteria ultimately cause disease is highly dependent upon understanding how they coexist and interact with one another in biofilm communities and the mechanisms by which biofilms are formed. Our research has demonstrated that the DNABII family of DNA-binding proteins are important components of the extracellular DNA (eDNA)-dependent matrix of bacterial biofilms and that sequestering these proteins via protein-specific antibodies results in the collapse of the biofilm structure and release of the resident bacteria. While the high degree of similarity among the DNABII family of proteins has allowed antibodies derived against specific DNABII proteins to disrupt biofilms formed by a wide range of bacterial pathogens, the DNABII proteins of P. gingivalis have proven to be antigenically distinct, allowing us to determine if we can use anti-P. gingivalis HUß antibodies to specifically target this species for removal from a mixed-species biofilm. Importantly, despite forming homotypic biofilms in vitro, P. gingivalis must enter preexisting biofilms in vivo in order to persist within the oral cavity. The data presented here indicate that antibodies derived against the P. gingivalis DNABII protein, HUß, reduce by half the amount of P. gingivalis organisms entering into preexisting biofilm formed by four oral streptococcal species. These results support our efforts to develop methods for preventing and treating periodontal disease.IMPORTANCE Periodontitis is one of the most prevalent chronic infections, affecting 40 to 50% of the population of the United States. The root cause of periodontitis is the presence of bacterial biofilms within the gingival space, with Porphyromonas gingivalis being strongly associated with the development of the disease. Periodontitis also increases the risk of secondary conditions and infections such as atherosclerosis and infective endocarditis caused by oral streptococci. To induce periodontitis, P. gingivalis needs to incorporate into preformed biofilms, with oral streptococci being important binding partners. Our research demonstrates that targeting DNABII proteins with an antibody disperses oral streptococcus biofilm and prevents P. gingivalis entry into oral streptococcus biofilm. These results suggest potential therapeutic treatments for endocarditis caused by streptococci as well as periodontitis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Bacteroidaceae/microbiología , Biopelículas/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Periodontitis/microbiología , Porphyromonas gingivalis/patogenicidad , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Proteínas de Unión al ADN/genética , Humanos , Boca/microbiología , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/crecimiento & desarrollo , Alineación de Secuencia
5.
J Biol Chem ; 291(24): 12538-12546, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27129225

RESUMEN

Biofilms are organized multicellular communities encased in an extracellular polymeric substance (EPS). Biofilm-resident bacteria resist immunity and antimicrobials. The EPS provides structural stability and presents a barrier; however, a complete understanding of how EPS structure relates to biological function is lacking. This review focuses on the EPS of three Gram-negative pathogens: Pseudomonas aeruginosa, nontypeable Haemophilus influenzae, and Salmonella enterica serovar Typhi/Typhimurium. Although EPS proteins and polysaccharides are diverse, common constituents include extracellular DNA, DNABII (DNA binding and bending) proteins, pili, flagella, and outer membrane vesicles. The EPS biochemistry promotes recalcitrance and informs the design of therapies to reduce or eliminate biofilm burden.


Asunto(s)
Infecciones Bacterianas/microbiología , Biopelículas/crecimiento & desarrollo , ADN Bacteriano/metabolismo , Matriz Extracelular/metabolismo , Bacterias Gramnegativas/fisiología , Polisacáridos Bacterianos/metabolismo , Antibacterianos/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Biopelículas/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Evasión Inmune/efectos de los fármacos , Evasión Inmune/fisiología
6.
Laryngoscope ; 134(4): 1564-1571, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37597166

RESUMEN

OBJECTIVES: We examined sinus mucosal samples recovered from pediatric chronic rhinosinusitis (CRS) patients for the presence of Z-form extracellular DNA (eDNA) due to its recently elucidated role in pathogenesis of disease. Further, we immunolabeled these specimens for the presence of both members of the bacterial DNA-binding DNABII protein family, integration host factor (IHF) and histone-like protein (HU), due to their known role in converting common B-DNA to the rare Z-form. METHODS: Sinus mucosa samples recovered from 20 patients during functional endoscopic sinus surgery (FESS) were immunolabelled for B- and Z-DNA, as well as for both bacterial DNABII proteins. RESULTS: Nineteen of 20 samples (95%) included areas rich in eDNA, with the majority in the Z-form. Areas positive for B-DNA were restricted to the most distal regions of the mucosal specimen. Labeling for both DNABII proteins was observed on B- and Z-DNA, which aligned with the role of these proteins in the B-to-Z DNA conversion. CONCLUSIONS: Abundant Z-form eDNA in culture-positive pediatric CRS samples suggested that bacterial DNABII proteins were responsible for the conversion of eukaryotic B-DNA that had been released into the luminal space by PMNs during NETosis, to the Z-form. The presence of both DNABII proteins on B-DNA and Z-DNA supported the known role of these bacterial proteins in the B-to-Z DNA conversion. Given that Z-form DNA both stabilizes the bacterial biofilm and inactivates PMN NET-mediated killing of trapped bacteria, we hypothesize that this conversion may be contributing to the chronicity and recalcitrance of CRS to treatment. LEVEL OF EVIDENCE: NA Laryngoscope, 134:1564-1571, 2024.


Asunto(s)
ADN Forma B , ADN de Forma Z , Rinitis , Sinusitis , Humanos , Niño , Factores de Integración del Huésped , Biopelículas , Sinusitis/cirugía , Enfermedad Crónica , Rinitis/cirugía
8.
Front Microbiol ; 14: 1202215, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37564292

RESUMEN

Introduction: The "silent" antimicrobial resistance (AMR) pandemic is responsible for nearly five million deaths annually, with a group of seven biofilm-forming pathogens, known as the ESKAPEE pathogens, responsible for 70% of these fatalities. Biofilm-resident bacteria, as they exist within the disease site, are canonically highly resistant to antibiotics. One strategy to counter AMR and improve disease resolution involves developing methods to disrupt biofilms. These methods aim to release bacteria from the protective biofilm matrix to facilitate their killing by antibiotics or immune effectors. Several laboratories working on such strategies have demonstrated that bacteria newly released from a biofilm display a transient phenotype of significantly increased susceptibility to antibiotics. Similarly, we developed an antibody-based approach for biofilm disruption directed against the two-membered DNABII family of bacterial DNA-binding proteins, which serve as linchpins to stabilize the biofilm matrix. The incubation of biofilms with α-DNABII antibodies rapidly collapses them to induce a population of newly released bacteria (NRel). Methods: In this study, we used a humanized monoclonal antibody (HuTipMab) directed against protective epitopes of a DNABII protein to determine if we could disrupt biofilms formed by the high-priority ESKAPEE pathogens as visualized by confocal laser scanning microscopy (CLSM) and COMSTAT2 analysis. Then, we demonstrated the potentiated killing of the induced NRel by seven diverse classes of traditional antibiotics by comparative plate count. Results: To this end, ESKAPEE biofilms were disrupted by 50%-79% using a single tested dose and treatment period with HuTipMab. The NRel of each biofilm were significantly more sensitive to killing than their planktonically grown counterparts (heretofore, considered to be the most sensitive to antibiotic-mediated killing), even when tested at a fraction of the MIC (1/250-1/2 MIC). Moreover, the bacteria that remained within the biofilms of two representative ESKAPEE pathogens after HuTipMab disruption were also significantly more susceptible to killing by antibiotics. Discussion: New data presented in this study support our continued development of a combinatorial therapy wherein HuTipMab is delivered to a patient with recalcitrant disease due to an ESKAPEE pathogen to disrupt a pathogenic biofilm, along with a co-delivered dose of an antibiotic whose ability to rapidly kill the induced NRel has been demonstrated. This novel regimen could provide a more successful clinical outcome to those with chronic, recurrent, or recalcitrant diseases, while limiting further contribution to AMR.

9.
Biofilm ; 6: 100166, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38078059

RESUMEN

Objectives: Structural or mucus hypersecretory pulmonary diseases such as cystic fibrosis (CF), wherein viscous mucus accumulates and clearance functions are impaired, predispose people to lung infection by inhaled bacteria that form biofilm aggregates. Nontuberculous mycobacteria (NTM), primarily Mycobacterium abscessus and Mycobacterium avium, are the growing cause of these lung infections and are extremely challenging to treat due to antibiotic recalcitrance. Better therapeutic approaches are urgently needed. We developed a humanized monoclonal antibody (HuTipMab) directed against a biofilm structural linchpin, the bacterial DNABII proteins, that rapidly disrupts biofilms and generates highly vulnerable newly released bacteria (NRel). Methods: HuTipMab's ability to recognize HupB, NTM's DNABII homologue was determined by ELISA. Relative ability of HuTipMab to disrupt biofilms formed by lab-passaged and clinical isolates of NTM was assessed by CLSM. Relative sensitivity of NTM NRel to antibiotic killing compared to when grown planktonically was evaluated by plate count. Results: HuTipMab recognized HupB and significantly disrupted NTM biofilms in a time- and dose-dependent manner. Importantly, NTM NRel of lab-passaged and clinical isolates were now highly sensitive to killing by amikacin and azithromycin. Conclusions: If successful, this combinatorial treatment strategy would empower existing antibiotics to more effectively kill NTM newly released from a biofilm by HuTipMab and thereby both improve clinical outcomes and perhaps decrease length of antibiotic treatment for people that are NTM culture-positive.

10.
Antibiotics (Basel) ; 11(1)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35052981

RESUMEN

Antimicrobial resistance (AMR) is a concerning global threat that, if not addressed, could lead to increases in morbidity and mortality, coupled with societal and financial burdens. The emergence of AMR bacteria can be attributed, in part, to the decreased development of new antibiotics, increased misuse and overuse of existing antibiotics, and inadequate treatment options for biofilms formed during bacterial infections. Biofilms are complex microbiomes enshrouded in a self-produced extracellular polymeric substance (EPS) that is a primary defense mechanism of the resident microorganisms against antimicrobial agents and the host immune system. In addition to the physical protective EPS barrier, biofilm-resident bacteria exhibit tolerance mechanisms enabling persistence and the establishment of recurrent infections. As current antibiotics and therapeutics are becoming less effective in combating AMR, new innovative technologies are needed to address the growing AMR threat. This perspective article highlights such a product, CMTX-101, a humanized monoclonal antibody that targets a universal component of bacterial biofilms, leading to pathogen-agnostic rapid biofilm collapse and engaging three modes of action-the sensitization of bacteria to antibiotics, host immune enablement, and the suppression of site-specific tissue inflammation. CMTX-101 is a new tool used to enhance the effectiveness of existing, relatively inexpensive first-line antibiotics to fight infections while promoting antimicrobial stewardship.

12.
Laryngoscope ; 131(10): E2698-E2704, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33666254

RESUMEN

OBJECTIVES/HYPOTHESIS: To evaluate the ability of humanized monoclonal antibody fragments directed against a bacterial DNABII protein plus ofloxacin delivered directly into the chinchilla middle ear via tympanostomy tube (TT) to enhance the ability of ofloxacin to eradicate biofilms formed by nontypeable Haemophilus influenzae (NTHI). STUDY DESIGN: A blinded pre-clinical study of comparative efficacy of single versus combinatorial treatment strategies. METHODS: NTHI was allowed to form biofilms in the middle ears of chinchillas prior to TT placement. Ofloxacin, humanized Fab fragments against a bacterial DNABII protein that disrupts biofilms or Fab fragments plus ofloxacin were instilled into the middle ear via TT. For two consecutive days, ofloxacin was delivered twice-a-day, Fab fragments were delivered once-a-day, or these treatments were combined. Relative biofilm resolution (as determined via two outcome measures) and eradication of viable NTHI were assessed 1-day later. RESULTS: Whereas ofloxacin alone did not resolve biofilms or eradicate NTHI from the middle ear, delivery of Fab fragments significantly reduced both biofilms and NTHI burden over this short course of treatment. Notably, co-delivery of ofloxacin plus humanized Fab fragments eradicated both NTHI and biofilms from the middle ear, an enhanced outcome compared to receipt of either treatment alone. CONCLUSION: This study demonstrated a powerful combinatorial approach to release bacteria from their protective biofilms and rapidly render them vulnerable to killing by a previously ineffective antibiotic. An approach to combine ofloxacin with humanized Fab fragments that disrupt biofilms has tremendous potential to quickly resolve chronic otorrhea suffered by children with chronic suppurative otitis media or chronic post-tympanostomy tube otorrhea and thereby improve their quality of life. LEVEL OF EVIDENCE: NA Laryngoscope, 131:E2698-E2704, 2021.


Asunto(s)
Biopelículas/efectos de los fármacos , Proteínas de Unión al ADN/farmacología , Infecciones por Haemophilus/tratamiento farmacológico , Fragmentos Fab de Inmunoglobulinas/farmacología , Ofloxacino/farmacología , Otitis Media/tratamiento farmacológico , Animales , Chinchilla , ADN Bacteriano , Haemophilus influenzae , Ventilación del Oído Medio
13.
Front Immunol ; 12: 656090, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841441

RESUMEN

Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with multiple autoantibody production and often affects the kidneys, known as lupus nephritis. However, the mechanism underlying lupus nephritis development is unclear. Biofilms that protect bacteria from stress are ubiquitous in almost every environment. Here, we identified that a conserved peptide (HU1) derived from DNABII proteins, one of major bacterial biofilm components, was specifically recognized by sera from about 47% patients with SLE. Moreover, the serum anti-HU1 levels showed a significant positive correlation with lupus nephritis occurrence. Presence of antibodies against HU1 in pristane-induced mice aggravated lupus nephritis, although these antibodies also attenuated bacterial biofilm formation. We further identified that antibodies against HU1 cross-recognized protein disulfide isomerase (P4HB) located on the renal cell surface and inhibited the activities of this enzyme. Our findings reveal a novel mechanism underlying the development of lupus nephritis triggered by bacterial biofilms.


Asunto(s)
Autoanticuerpos/inmunología , Bacterias/inmunología , Biopelículas , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/etiología , Nefritis Lúpica/patología , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Autoanticuerpos/sangre , Autoantígenos/inmunología , Biomarcadores , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/sangre , Ratones , Ratones Transgénicos , Péptidos/química , Péptidos/inmunología , Terpenos/efectos adversos
14.
mSphere ; 5(2)2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32295873

RESUMEN

The use of broad-spectrum antibiotics to treat diseases, such as the highly prevalent pediatric disease otitis media (OM), contributes significantly to the worldwide emergence of multiple-antibiotic-resistant microbes, and gut dysbiosis with diarrhea is a common adverse sequela. Moreover, for many diseases, like OM, biofilms contribute significantly to chronicity and recurrence, yet biofilm-resident bacteria are characteristically highly resistant to antibiotics. The most cost-effective way to both prevent and resolve diseases like OM, as well as begin to address the problem of growing antibiotic resistance, would be via the development of novel approaches to eradicate bacterial biofilms. Toward this goal, we designed a vaccine antigen that induces the formation of antibodies that prevent biofilm formation and, thereby, experimental OM in the middle ears of chinchillas by the predominant Gram-negative pathogen responsible for this disease, nontypeable Haemophilus influenzae These antibodies also significantly disrupt preexisting biofilms formed by diverse pathogens. Whereas preclinical data strongly support the continued development of this vaccine antigen, which targets an essential structural element of bacterial biofilms, a concern has been whether active immunization would also lead to unintended collateral damage in the form of an altered gut microbiome. To address this concern, we assessed changes in the microbiome of the chinchilla gut over time after the delivery of either amoxicillin-clavulanate, the standard of care for OM, or after immunization with our biofilm-targeted vaccine antigen either via a traditional subcutaneous route or via a novel noninvasive transcutaneous route. We show that differences in the abundance of specific taxa were found only in the stools of antibiotic-treated animals.IMPORTANCE The prevalence of chronic and recurrent diseases, combined with the overuse/abuse of antibiotics that has led to the sobering emergence of bacteria resistant to multiple antibiotics, has mandated that we develop novel approaches to better manage these diseases or, ideally, prevent them. Biofilms play a key role in the pathogenesis of chronic and recurrent bacterial diseases but are difficult, if not impossible, to eradicate with antibiotics. We developed a vaccine antigen designed to mediate biofilm disruption; however, it is also important that delivery of this vaccine does not induce collateral damage to the microbiome. The studies described here validated a vaccine approach that targets biofilms without the consequences of an altered gut microbiome. While delivery of the antibiotic most commonly given to children with ear infections did indeed alter the gut microbiome, as expected, immunization via traditional injection or by noninvasive delivery to the skin did not result in changes to the chinchilla gut microbiome.


Asunto(s)
Antígenos Bacterianos/administración & dosificación , Biopelículas/crecimiento & desarrollo , Microbioma Gastrointestinal , Vacunas contra Haemophilus/administración & dosificación , Otitis Media/prevención & control , Administración Oral , Combinación Amoxicilina-Clavulanato de Potasio , Animales , Antibacterianos , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Chinchilla/microbiología , Estudios de Cohortes , Oído Medio/microbiología , Femenino , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/prevención & control , Haemophilus influenzae/inmunología , Haemophilus influenzae/patogenicidad , Inmunización , Masculino , Otitis Media/tratamiento farmacológico , Otitis Media/microbiología
15.
Expert Opin Ther Pat ; 30(12): 907-909, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32970473

RESUMEN

INTRODUCTION: To date, microbial infections are also difficult to eradicate due to the increasing capability of bacteria to form a biofilm. In the era of antibiotic resistance, antibody-based approaches represent great promise in curing infective pathogens. The authors of US2019338018 patent propose a method for the treatment of biofilm-related disorders by using specific antibody fragments. AREAS COVERED: The US2019338018 patent reports antibody fragments, pharmaceutical composition that contains it, and their application for the treatment of biofilm-linked disorders. Proof concept and preclinical results show that mAb mIhfB4NTHI Fab caused robust eradication of the biofilm in the middle ear lumen of chinchillas affected by Hemophilus influenzae infection. EXPERT OPINION: Fab fragments of the US2019338018 patent are new in a general concept to treat bacterial biofilms and biofilm-linked disorders. However, pre-clinical data are only shown for the treatment with Fab fragments of infections caused by H. influenzae in the middle ear of chinchillas. There are no clinical trials that demonstrate that the treatment with Fab fragments may induce a disruption of biofilm produced by H. influenzae or other pathogens and an anti-inflammatory response in infected patients.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Infecciones por Haemophilus/tratamiento farmacológico , Fragmentos de Inmunoglobulinas/inmunología , Animales , Chinchilla , Oído Medio/efectos de los fármacos , Oído Medio/microbiología , Infecciones por Haemophilus/inmunología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Patentes como Asunto
16.
Laryngoscope ; 130(3): 806-811, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31021431

RESUMEN

OBJECTIVES: Otitis media with effusion (OME) is a common disease of childhood that is largely asymptomatic. However, middle ear fluid can persist for months and negatively impact a child's quality of life. Many cases of OME remain chronic and require surgical intervention. Because biofilms are known to contribute to the persistence of many diseases, this study examined effusions collected from children with chronic OME for the presence of essential biofilm structural components, members of the DNABII family of bacterial DNA-binding proteins. METHODS: Middle ear effusions were recovered from 38 children with chronic OME at the time of tympanostomy tube insertion. A portion of each specimen was submitted for microbiology culture. The remaining material was assessed by immunoblot to quantitate individual DNABII proteins, integration host factor (IHF), and histone-like protein (HU). RESULTS: Sixty-five percent of effusions (24 of 37) were culture-positive for bacterial species or yeast, whereas 35% (13 of 37) were culture-negative. IHF was detected in 95% (36 of 38) at concentrations from 2 to 481 ng/µL effusion. HU was detected in 95% (36 of 38) and quantitated from 13 to 5,264 ng/µL effusion (P ≤ 0.05 compared to IHF). CONCLUSION: Because DNABII proteins are essential structural components of bacterial biofilms, these data lend further support to our understanding that biofilms are present in the vast majority of chronic middle ear effusions, despite negative culture results. The presence and ubiquity of DNABII proteins in OME specimens indicated that these proteins can serve as an important clinical target for our novel DNABII-directed strategy to treat biofilm diseases such as chronic OME. LEVEL OF EVIDENCE: NA Laryngoscope, 130:806-811, 2020.


Asunto(s)
Proteínas Bacterianas/análisis , Biopelículas , Exudados y Transudados/química , Otitis Media con Derrame , Adolescente , Estructuras Bacterianas , Niño , Preescolar , Exudados y Transudados/microbiología , Femenino , Humanos , Lactante , Masculino , Otitis Media con Derrame/microbiología , Adulto Joven
17.
J Pediatr Infect Dis ; 14(2): 69-77, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30853830

RESUMEN

Otitis media (OM) is one of the most common diseases of childhood, and nontypeable Haemophilus influenzae (NTHI) is the predominant causative agent of chronic and recurrent OM, as well as OM for which treatment has failed. Moreover, NTHI is now as important a causative agent of acute OM as the pneumococcus. NTHI colonizes the human nasopharynx asymptomatically. However, upon perturbation of the innate and physical defenses of the airway by upper respiratory tract viral infection, NTHI can replicate, ascend the Eustachian tube, gain access to the normally sterile middle ear space, and cause disease. Bacterial biofilms within the middle ear, including those formed by NTHI, contribute to the chronic and recurrent nature of this disease. These multicomponent structures are highly resistant to clearance by host defenses and elimination by traditional antimicrobial therapies. Herein, we review several strategies utilized by NTHI in order to persist within the human host and interventions currently under investigation to prevent and/or resolve NTHI-induced diseases of the middle ear and uppermost airway.

18.
mBio ; 9(5)2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30228240

RESUMEN

Biofilms play a critical role in the colonization, persistence, and pathogenesis of many human pathogens. Multiple mucosa-associated pathogens have evolved a mechanism of rapid adaptation, termed the phasevarion, which facilitates a coordinated regulation of numerous genes throughout the bacterial genome. This epigenetic regulation occurs via phase variation of a DNA methyltransferase, Mod. The phasevarion of nontypeable Haemophilus influenzae (NTHI) significantly affects the severity of experimental otitis media and regulates several disease-related processes. However, the role of the NTHI phasevarion in biofilm formation is unclear. The present study shows that the phasevarions of multiple NTHI clinical isolates regulate in vitro biofilm formation under disease-specific microenvironmental conditions. The impact of phasevarion regulation was greatest under alkaline conditions that mimic those known to occur in the middle ear during disease. Under alkaline conditions, NTHI strains that express the ModA2 methyltransferase formed biofilms with significantly greater biomass and less distinct architecture than those formed by a ModA2-deficient population. The biofilms formed by NTHI strains that express ModA2 also contained less extracellular DNA (eDNA) and significantly less extracellular HU, a DNABII DNA-binding protein critical for biofilm structural stability. Stable biofilm structure is critical for bacterial pathogenesis and persistence in multiple experimental models of disease. These results identify a role for the phasevarion in regulation of biofilm formation, a process integral to the chronic nature of many infections. Understanding the role of the phasevarion in biofilm formation is critical to the development of prevention and treatment strategies for these chronic diseases.IMPORTANCE Upper respiratory tract infections are the number one reason for a child to visit the emergency department, and otitis media (middle ear infection) ranks third overall. Biofilms contribute significantly to the chronic nature of bacterial respiratory tract infections, including otitis media, and make these diseases particularly difficult to treat. Several mucosa-associated human pathogens utilize a mechanism of rapid adaptation termed the phasevarion, or phasevariable regulon, to resist environmental and host immune pressures. In this study, we assessed the role of the phasevarion in regulation of biofilm formation by nontypeable Haemophilus influenzae (NTHI), which causes numerous respiratory tract diseases. We found that the NTHI phasevarion regulates biofilm structure and critical biofilm matrix components under disease-specific conditions. The findings of this work could be significant in the design of improved strategies against NTHI infections, as well as diseases due to other pathogens that utilize a phasevarion.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Epigénesis Genética , Regulación Bacteriana de la Expresión Génica , Haemophilus influenzae/genética , Álcalis , Infecciones por Haemophilus/microbiología , Humanos , Concentración de Iones de Hidrógeno , Otitis Media/microbiología , Fenotipo , Regulón/genética
19.
Mol Oral Microbiol ; 32(1): 74-88, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-26931773

RESUMEN

Periodontal disease exemplifies a chronic and recurrent infection with a necessary biofilm component. Mucosal inflammation is a hallmark response of the host seen in chronic diseases, such as colitis, gingivitis, and periodontitis (and the related disorder peri-implantitis). We have taken advantage of our recently developed rat model of human peri-implantitis that recapitulates osteolysis, the requirement of biofilm formation, and the perpetuation of the bona fide disease state, to test a new therapeutic modality with two novel components. First we used hyperimmune antiserum directed against the DNABII family of proteins, now known to be a critical component of the extracellular matrix of bacterial biofilms. Second we delivered the antiserum as cargo in biodegradable microspheres to the site of the biofilm infection. We demonstrated that delivery of a single dose of anti-DNABII in poly(lactic-co-glycolic acid) (PLGA) microspheres induced significant resolution of experimental peri-implantitis, including marked reduction of inflammation. These data support the continued development of a DNABII protein-targeted therapeutic for peri-implantitis and other chronic inflammatory pathologies of the oral cavity in animals and humans.


Asunto(s)
Biopelículas/efectos de los fármacos , Proteínas de Unión al ADN/inmunología , Osteólisis/inmunología , Osteólisis/microbiología , Osteólisis/terapia , Periodontitis/microbiología , Animales , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Biopelículas/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Implantes Dentales/microbiología , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/inmunología , Femenino , Factores de Integración del Huésped/inmunología , Ácido Láctico/farmacología , Microesferas , Osteólisis/patología , Periimplantitis/inmunología , Periimplantitis/microbiología , Periimplantitis/patología , Periimplantitis/terapia , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Conejos , Ratas , Ratas Sprague-Dawley
20.
Mol Oral Microbiol ; 32(2): 118-130, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26988714

RESUMEN

Bacteria that persist in the oral cavity exist within complex biofilm communities. A hallmark of biofilms is the presence of an extracellular polymeric substance (EPS), which consists of polysaccharides, extracellular DNA (eDNA), and proteins, including the DNABII family of proteins. The removal of DNABII proteins from a biofilm results in the loss of structural integrity of the eDNA and the collapse of the biofilm structure. We examined the role of DNABII proteins in the biofilm structure of the periodontal pathogen Porphyromonas gingivalis and the oral commensal Streptococcus gordonii. Co-aggregation with oral streptococci is thought to facilitate the establishment of P. gingivalis within the biofilm community. We demonstrate that DNABII proteins are present in the EPS of both S. gordonii and P. gingivalis biofilms, and that these biofilms can be disrupted through the addition of antisera derived against their respective DNABII proteins. We provide evidence that both eDNA and DNABII proteins are limiting in S. gordonii but not in P. gingivalis biofilms. In addition, these proteins are capable of complementing one another functionally. We also found that whereas antisera derived against most DNABII proteins are capable of binding a wide variety of DNABII proteins, the P. gingivalis DNABII proteins are antigenically distinct. The presence of DNABII proteins in the EPS of these biofilms and the antigenic uniqueness of the P. gingivalis proteins provide an opportunity to develop therapies that are targeted to remove P. gingivalis and biofilms that contain P. gingivalis from the oral cavity.


Asunto(s)
Antígenos Bacterianos/inmunología , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/inmunología , Biopelículas , Proteínas de Unión al ADN/inmunología , AdnB Helicasas/inmunología , Porphyromonas gingivalis/fisiología , Streptococcus gordonii/fisiología , Anticuerpos Antibacterianos/inmunología , ADN Bacteriano/metabolismo , Microscopía Fluorescente , Porphyromonas gingivalis/efectos de los fármacos , Porphyromonas gingivalis/inmunología , Streptococcus gordonii/efectos de los fármacos , Streptococcus gordonii/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA