Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuromodulation ; 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-38165292

RESUMO

OBJECTIVE: This long-term retrospective study evaluated the survival and amputation outcome of subjects who received neuromodulation therapy for the management of peripheral arterial disease (PAD). MATERIALS AND METHODS: The study reviews the health data of a single cohort of 51 patients who received spinal neuromodulation (spinal cord stimulation [SCS] or dorsal root ganglion stimulation [DRG-S]) for PAD from 2007 to 2022 in a single German center. Survival rate and major amputation rate were determined. Pain, quality of life, walking distance, and opioid usage were assessed before implantation (baseline), one, six, and 12 months (M) after implantation, and then annually (during a follow-up visit). Implant-related complications also were documented. RESULTS: In total, 51 patients (37 men [mean age 68.9 ± 10.2 years], 14 women [mean age (68.7 ± 14.6 years]) underwent SCS (n = 49) or DRG-S (n = 2) implantation owing to persistent ischemic pain. The follow-up mean years ± SD is 4.04 ± 2.73. At baseline, patients were classified as Rutherford's category 3 (n = 23), category 4 (n = 15) or category 5 (n = 9). At 24 M, 42 of 47 patients did not require a major amputation after the implant. All the patients reported nearly complete pain relief from pain at rest. A total of 75% of patients were able to walk >200 m, and 87% of patients who used opioids at baseline were off this medication at 24 M. Overall, 93% of patients reported an improvement in their overall health assessment. These improved outcomes were sustained through years three to 10 for patients who have reported outcomes. CONCLUSIONS: Our single-center data support the efficacy of spinal neuromodulation for improvements in limb salvage, pain relief, mobility, and quality of life. The data also show that neuromodulative therapy has a long-term therapeutic effect in patients with chronic limb pain with Rutherford category 3, 4, and 5 PAD.

2.
Microbiology (Reading) ; 168(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35293855

RESUMO

Type IV pili are involved in adhesion, twitching motility, aggregation, biofilm formation and virulence in a variety of Gram-negative bacteria. Burkholderia pseudomallei, the causative agent of melioidosis and a Tier 1 biological select agent, is a Gram-negative bacterium with eight type IV pili-associated loci (TFP1 to TFP8). Most have not been fully characterized. In this study, we investigated BPSS2185, an uncharacterized TFP8 gene that encodes a type IVB pilus protein subunit. Using genetic deletion and complementation analysis in B. pseudomallei JW270, we demonstrate that BPSS2185 plays an important role in twitching motility and adhesion to A549 human alveolar epithelial cells. Compared to JW270, the JW270 ΔBPSS2185 mutant failed to display twitching motility and did not adhere to the epithelial cells. These phenotypes were partially reversed by the complementation of BPSS2185 in the mutant strain. The study also shows that BPSS2185 is expressed only during the onset of mature biofilm formation and at the dispersal of a biofilm, suggesting that the motility characteristic is required to form a biofilm. Our study is the first to suggest that the BPSS2185 gene in TFP8 contributes to twitching motility, adhesion and biofilm formation, indicating that the gene may contribute to B. pseudomallei virulence.


Assuntos
Burkholderia pseudomallei , Proteínas de Fímbrias , Biofilmes , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Células Epiteliais/metabolismo , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo
3.
J Pain Res ; 14: 3259-3265, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34703301

RESUMO

BACKGROUND: Chronic pain patients implanted with a neurostimulation device typically require follow-up and device programming visits to address changes in symptoms or treatment. Follow-up visits require access to specialty care and necessitate patients to take time off work, commute long distances, arrange for travel, and/or work with a caregiver's schedule. Telemedicine was adopted for some patient management as a result of the Sars-Cov-2 pandemic; however, remote optimization for neuromodulation still required an in-person visit to adjust device parameters. An FDA-approved digital platform enables remote programming of an implanted neuromodulation device using a real-time audio-video link from the clinical programmer to the patient controller. The Remote Optimization, Adjustment, and Measurement for Chronic Pain Therapy (ROAM-CPT) is a multi-center, prospective study that is currently underway to access the effectiveness of the teleprogramming system in fulfilling patients' clinical demands. METHODS: This pilot study surveyed 16 patients to determine the ability of the teleprogramming platform to provide a rapid solution safely and effectively for patient's chronic pain. Data were collected using a questionnaire that asked 6 clinician-centric questions and 5 patient-centric questions. RESULTS: 4/4 surveyed physicians were able to address patients' needs. 16/16 surveyed patients reported a quick resolution to pain and 15/16 did not require additional follow-up. Data curated from this pilot study show that the teleprogramming application greatly improves patient care, is preferred by both clinicians and patients with minimal disruptions to patients' everyday lives. CONCLUSION: Teleprogramming provides real-time virtual programming capabilities and optimizes patients' therapy. PERSPECTIVE: This article describes remote device programming and analysis as an alternative to in-person programming/treatment sessions for neuromodulation patients. This remote option gives patients access to timely and clinically appropriate device management when in-person care may not be available.

4.
Microbiol Spectr ; 9(1): e0010221, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34160272

RESUMO

Burkholderia pseudomallei is an opportunistic pathogen that is responsible for the disease melioidosis in humans and animals. The microbe is a tier 1 select agent because it is highly infectious by the aerosol route, it is inherently resistant to multiple antibiotics, and no licensed vaccine currently exists. Naturally acquired infections result from contact with contaminated soil or water sources in regions of endemicity. There have been few reports investigating the molecular mechanism(s) utilized by B. pseudomallei to survive and persist in ecological niches harboring microbial competitors. Here, we report the isolation of Gram-positive bacteria from multiple environmental sources and show that ∼45% of these isolates are inhibited by B. pseudomallei in head-to-head competition assays. Two competition-deficient B. pseudomallei transposon mutants were identified that contained insertion mutations in the hmqA-G operon. This large biosynthetic gene cluster encodes the enzymes that produce a family of secondary metabolites called 4-hydroxy-3-methyl-2-alkylquinolines (HMAQs). Liquid chromatography and mass spectrometry conducted on filter-sterilized culture supernatants revealed five HMAQs and N-oxide derivatives that were produced by the parental strain but were absent in an isogenic hmqD deletion mutant. The results demonstrate that B. pseudomallei inhibits the growth of environmental Gram-positive bacteria in a contact-independent manner via the production of HMAQs by the hmqA-G operon. IMPORTANCE Burkholderia pseudomallei naturally resides in water, soil, and the rhizosphere and its success as an opportunistic pathogen is dependent on the ability to persist in these harsh habitats long enough to come into contact with a susceptible host. In addition to adapting to limiting nutrients and diverse chemical and physical challenges, B. pseudomallei also has to interact with a variety of microbial competitors. Our research shows that one of the ways in which B. pseudomallei competes with Gram-positive environmental bacteria is by exporting a diverse array of closely related antimicrobial secondary metabolites.


Assuntos
Proteínas de Bactérias/genética , Burkholderia pseudomallei/fisiologia , Bactérias Gram-Positivas/fisiologia , Interações Microbianas , Antibacterianos/biossíntese , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Burkholderia pseudomallei/genética , Bactérias Gram-Positivas/efeitos dos fármacos , Mutagênese Insercional , Óperon , Metabolismo Secundário
5.
Microorganisms ; 9(4)2021 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-33919891

RESUMO

Bartonella henselae (B. henselae) is a gram-negative bacterium that causes cat scratch disease, bacteremia, and endocarditis, as well as other clinical presentations. B. henselae has been shown to form a biofilm in vitro that likely plays a role in the establishment and persistence of the bacterium in the host. Biofilms are also known to form in the cat flea vector; hence, the ability of this bacterium to form a biofilm has broad biological significance. The release of B. henselae from a biofilm niche appears to be important in disease persistence and relapse in the vertebrate host but also in transmission by the cat flea vector. It has been shown that the BadA adhesin of B. henselae is critical for adherence and biofilm formation. Thus, the upregulation of badA is important in initiating biofilm formation, and down-regulation is important in the release of the bacterium from the biofilm. We summarize the current knowledge of biofilm formation in Bartonella species and the role of BadA in biofilm formation. We discuss the evidence that defines possible mechanisms for the regulation of the genes required for biofilm formation. We further describe the regulation of those genes in the conditions that mimic both the arthropod vector and the mammalian host for B. henselae. The treatment for persistent B. henselae infection remains a challenge; hence, a better understanding of the mechanisms by which this bacterium persists in its host is critical to inform future efforts to develop drugs to treat such infections.

6.
Virulence ; 11(1): 1041-1058, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32835600

RESUMO

Burkholderia pseudomallei: is the etiological agent of the disease melioidosis and is a Tier 1 select agent. It survives and replicates inside phagocytic cells by escaping from the endocytic vacuole, replicating in the cytosol, spreading to other cells via actin polymerization and promoting the fusion of infected and uninfected host cells to form multinucleated giant cells. In this study, we utilized a proteomics approach to identify bacterial proteins produced inside RAW264.7 murine macrophages and host proteins produced in response to B. pseudomallei infection. Cells infected with B. pseudomallei strain K96243 were lysed and the lysate proteins digested and analyzed using nanoflow reversed-phase liquid chromatography and tandem mass spectrometry. Approximately 160 bacterial proteins were identified in the infected macrophages, including BimA, TssA, TssB, Hcp1 and TssM. Several previously uncharacterized B. pseudomallei proteins were also identified, including BPSS1996 and BPSL2748. Mutations were constructed in the genes encoding these novel proteins and their relative virulence was assessed in BALB/c mice. The 50% lethal dose for the BPSS1996 mutant was approximately 55-fold higher than that of the wild type, suggesting that BPSS1996 is required for full virulence. Sera from B. pseudomallei-infected animals reacted with BPSS1996 and it was found to localize to the bacterial surface using indirect immunofluorescence. Finally, we identified 274 host proteins that were exclusively present or absent in infected RAW264.7 cells, including chemokines and cytokines involved in controlling the initial stages of infection.


Assuntos
Proteínas de Bactérias/imunologia , Burkholderia pseudomallei/patogenicidade , Fatores de Virulência/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Feminino , Macaca mulatta , Macrófagos/imunologia , Macrófagos/microbiologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Virulência , Fatores de Virulência/genética
7.
Microb Pathog ; 147: 104272, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32464301

RESUMO

Bartonella henselae (Bh) is a Gram-negative zoonotic bacterium that can grow as large aggregates and form biofilms in vitro dependent upon the adhesin BadA. Previously, we reported that the Houston-1 strain of Bh has a family of nine small, highly-expressed intergenic transcripts called Bartonellaregulatory transcripts, Brt1-9. Each of the Brts bears a stem and loop structure on the 3' end followed by a gene encoding a DNA binding protein called the Transcriptional regulatory proteins, Trp1-9. RNA-seq analysis of laboratory-grown bacteria revealed the trps were poorly transcribed suggesting that the 3' stem and loop on the Brts results in transcript termination upstream of the trp genes under these conditions. Here we demonstrate that transcription of brt1 continues into trp1 when Bh is grown in a biofilm. Deletion of brt1, or just the 3' terminus of brt1 (containing the stem and loop structure), resulted in increased transcription of both trp1 and badA and increased biofilm formation. Trp1 was shown to directly bind the putative badA promoter region as demonstrated by an electrophoretic mobility shift assay (EMSA). Our data suggest that the 3' end of brt1 responds to a stimulus generated by growth of Bh in an in vitro biofilm to allow increased trp1 transcription. We further show that transcription of trp1 increases under conditions consistent with the mammalian host but is not highly expressed in the cat flea vector until the bacterium is excreted into the flea feces. Based on these data, we hypothesize that the 3' end of Brt1 functions to control trp1 transcription and Trp1 in turn results in increased badA expression and enhanced biofilm formation.


Assuntos
Bartonella henselae , Adesinas Bacterianas , Animais , Bartonella henselae/genética , Biofilmes , Proteínas de Ligação a DNA , RNA não Traduzido
8.
NPJ Biofilms Microbiomes ; 5(1): 10, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30886729

RESUMO

Bartonella henselae (Bh) is a Gram-negative rod transmitted to humans by a scratch from the common house cat. Infection of humans with Bh can result in a range of clinical diseases including lymphadenopathy observed in cat-scratch disease and more serious disease from persistent bacteremia. It is a common cause of blood-culture negative endocarditis as the bacterium is capable of growing as aggregates, and forming biofilms on infected native and prosthetic heart valves. The aggregative growth requires a trimeric autotransporter adhesin (TAA) called Bartonella adhesin A (BadA). TAAs are found in all Bartonella species and many other Gram-negative bacteria. Using Bh Houston-1, Bh Houston-1 ∆badA and Bh Houston-1 ∆badA/pNS2PTrc badA (a partial complement of badA coding for a truncated protein of 741 amino acid residues), we analyze the role of BadA in adhesion and biofilm formation. We also investigate the role of environmental factors such as temperature on badA expression and biofilm formation. Real-time cell adhesion monitoring and electron microscopy show that Bh Houston-1 adheres and forms biofilm more efficiently than the Bh Houston-1 ∆badA. Deletion of the badA gene significantly decreases adhesion, the first step in biofilm formation in vitro, which is partially restored in Bh Houston-1 ∆badA/pNS2PTrc badA. The biofilm formed by Bh Houston-1 includes polysaccharides, proteins, and DNA components and is susceptible to enzymatic degradation of these components. Furthermore, both pH and temperature influence both badA expression and biofilm formation. We conclude that BadA is required for optimal adhesion, agglutination and biofilm formation.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Bartonella henselae/crescimento & desenvolvimento , Biofilmes/crescimento & desenvolvimento , Fatores de Virulência/metabolismo , Bartonella henselae/genética , Bartonella henselae/efeitos da radiação , Biofilmes/efeitos da radiação , Deleção de Genes , Teste de Complementação Genética , Concentração de Íons de Hidrogênio , Temperatura , Fatores de Virulência/deficiência
9.
Clin Microbiol Rev ; 30(3): 709-746, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28490579

RESUMO

Since the reclassification of the genus Bartonella in 1993, the number of species has grown from 1 to 45 currently designated members. Likewise, the association of different Bartonella species with human disease continues to grow, as does the range of clinical presentations associated with these bacteria. Among these, blood-culture-negative endocarditis stands out as a common, often undiagnosed, clinical presentation of infection with several different Bartonella species. The limitations of laboratory tests resulting in this underdiagnosis of Bartonella endocarditis are discussed. The varied clinical picture of Bartonella infection and a review of clinical aspects of endocarditis caused by Bartonella are presented. We also summarize the current knowledge of the molecular basis of Bartonella pathogenesis, focusing on surface adhesins in the two Bartonella species that most commonly cause endocarditis, B. henselae and B. quintana. We discuss evidence that surface adhesins are important factors for autoaggregation and biofilm formation by Bartonella species. Finally, we propose that biofilm formation is a critical step in the formation of vegetative masses during Bartonella-mediated endocarditis and represents a potential reservoir for persistence by these bacteria.


Assuntos
Infecções por Bartonella/microbiologia , Bartonella/fisiologia , Endocardite/microbiologia , Infecções por Bartonella/sangue , Infecções por Bartonella/diagnóstico , Infecções por Bartonella/patologia , Endocardite/sangue , Endocardite/diagnóstico , Endocardite/patologia , Humanos
10.
Microbiologyopen ; 6(2)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27790856

RESUMO

Bartonella henselae is a gram-negative zoonotic bacterium that causes infections in humans including endocarditis and bacillary angiomatosis. B. henselae has been shown to grow as large aggregates and form biofilms in vitro. The aggregative growth and the angiogenic host response requires the trimeric autotransporter adhesin BadA. We examined the transcriptome of the Houston-1 strain of B. henselae using RNA-seq revealing nine novel, highly-expressed intergenic transcripts (Bartonella regulatory transcript, Brt1-9). The Brt family of RNAs is unique to the genus Bartonella and ranges from 194 to 203 nucleotides with high homology and stable predicted secondary structures. Immediately downstream of each of the nine RNA genes is a helix-turn-helix DNA-binding protein (transcriptional regulatory protein, Trp1-9) that is poorly transcribed under the growth conditions used for RNA-seq. Using knockdown or overexpressing strains, we show a role of both the Brt1 and Trp1 in the regulation of badA and also in biofilm formation. Based on these data, we hypothesize that Brt1 is a trans-acting sRNA that also serves as a cis-acting riboswitch to control the expression of badA. This family of RNAs together with the downstream Trp DNA-binding proteins represents a novel coordinated regulatory circuit controlling expression of virulence-associated genes in the bartonellae.


Assuntos
Angiomatose Bacilar/microbiologia , Bartonella henselae/genética , Bartonella henselae/patogenicidade , RNA Bacteriano/genética , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Bartonella henselae/isolamento & purificação , Sequência de Bases , Ctenocephalides/microbiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Técnicas de Silenciamento de Genes , Genoma Bacteriano/genética , Humanos , Proteínas de Membrana Transportadoras/genética , Análise de Sequência de RNA , Transcriptoma/genética , Fatores de Virulência/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA