301
|
Gong Q, Ali T, Hu Y, Gao R, Mou S, Luo Y, Yang C, Li A, Li T, Hao LL, He L, Yu X, Li S. RIPK1 inhibition mitigates neuroinflammation and rescues depressive-like behaviors in a mouse model of LPS-induced depression. Cell Commun Signal 2024; 22:427. [PMID: 39223674 PMCID: PMC11367892 DOI: 10.1186/s12964-024-01796-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Depression is often linked to inflammation in the brain. Researchers have been exploring ways to reduce this inflammation to improve depression symptoms. One potential target is a protein called RIPK1, which is known to contribute to brain inflammation. However, it's unclear how RIPK1 influences depression. Our study aims to determine whether RIPK1 inhibition could alleviate neuroinflammation-associated depression and elucidate its underlying mechanisms. METHODS To investigate our research objectives, we established a neuroinflammation mouse model by administering LPS. Behavioral and biochemical assessments were conducted on these mice. The findings were subsequently validated through in vitro experiments. RESULTS Using LPS-induced depression models, we investigated RIPK1's role, observing depressive-like behaviors accompanied by elevated cytokines, IBA-1, GFAP levels, and increased inflammatory signaling molecules and NO/H2O2. Remarkably, Necrostatin (Nec-1 S), a RIPK1 inhibitor, mitigated these changes. We further found altered expression and phosphorylation of eIF4E, PI3K/AKT/mTOR, and synaptic proteins in hippocampal tissues, BV2, and N2a cells post-LPS treatment, which Nec-1 S also ameliorated. Importantly, eIF4E inhibition reversed some of the beneficial effects of Nec-1 S, suggesting a complex interaction between RIPK1 and eIF4E in LPS-induced neuroinflammation. Moreover, citronellol, a RIPK1 agonist, significantly altered eIF4E phosphorylation, indicating RIPK1's potential upstream regulatory role in eIF4E and its contribution to neuroinflammation-associated depression. CONCLUSION These findings propose RIPK1 as a pivotal mediator in regulating neuroinflammation and neural plasticity, highlighting its significance as a potential therapeutic target for depression.
Collapse
Affiliation(s)
- Qichao Gong
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yue Hu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Ruyan Gao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Shengnan Mou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yanhua Luo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Canyu Yang
- College of Forensic Medicine, Institute of Forensic Injury, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi, China
| | - Axiang Li
- College of Forensic Medicine, Institute of Forensic Injury, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi, China
| | - Tao Li
- College of Forensic Medicine, Institute of Forensic Injury, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi, China
| | - Liang Liang Hao
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-Qiao Road, Chengdu, P.R. China
| | - Liufang He
- Department of Neonatology, Affiliated Longhua People's Hospital, Southern Medical University (Longhua People's Hospital), Shenzhen, 518190, China.
| | - Xiaoming Yu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People's Republic of China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
302
|
Sarkar R, Choudhury SM, Kanneganti TD. Classical apoptotic stimulus, staurosporine, induces lytic inflammatory cell death, PANoptosis. J Biol Chem 2024; 300:107676. [PMID: 39151726 PMCID: PMC11418131 DOI: 10.1016/j.jbc.2024.107676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/27/2024] [Accepted: 08/03/2024] [Indexed: 08/19/2024] Open
Abstract
Innate immunity is the body's first line of defense against disease, and regulated cell death is a central component of this response that balances pathogen clearance and inflammation. Cell death pathways are generally categorized as non-lytic and lytic. While non-lytic apoptosis has been extensively studied in health and disease, lytic cell death pathways are also increasingly implicated in infectious and inflammatory diseases and cancers. Staurosporine (STS) is a well-known inducer of non-lytic apoptosis. However, in this study, we observed that STS also induces lytic cell death at later timepoints. Using biochemical assessments with genetic knockouts, pharmacological inhibitors, and gene silencing, we identified that STS triggered PANoptosis via the caspase-8/RIPK3 axis, which was mediated by RIPK1. PANoptosis is a lytic, innate immune cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosome complexes. Deletion of caspase-8 and RIPK3, core components of the PANoptosome complex, protected against STS-induced lytic cell death. Overall, our study identifies STS as a time-dependent inducer of lytic cell death, PANoptosis. These findings emphasize the importance of understanding trigger- and time-specific activation of distinct cell death pathways to advance our understanding of the molecular mechanisms of innate immunity and cell death for clinical translation.
Collapse
Affiliation(s)
- Roman Sarkar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sk Mohiuddin Choudhury
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
303
|
Emamnejad R, Pagnin M, Petratos S. The iron maiden: Oligodendroglial metabolic dysfunction in multiple sclerosis and mitochondrial signaling. Neurosci Biobehav Rev 2024; 164:105788. [PMID: 38950685 DOI: 10.1016/j.neubiorev.2024.105788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease, governed by oligodendrocyte (OL) dystrophy and central nervous system (CNS) demyelination manifesting variable neurological impairments. Mitochondrial mechanisms may drive myelin biogenesis maintaining the axo-glial unit according to dynamic requisite demands imposed by the axons they ensheath. The promotion of OL maturation and myelination by actively transporting thyroid hormone (TH) into the CNS and thereby facilitating key transcriptional and metabolic pathways that regulate myelin biogenesis is fundamental to sustain the profound energy demands at each axo-glial interface. Deficits in regulatory functions exerted through TH for these physiological roles to be orchestrated by mature OLs, can occur in genetic and acquired myelin disorders, whereby mitochondrial efficiency and eventual dysfunction can lead to profound oligodendrocytopathy, demyelination and neurodegenerative sequelae. TH-dependent transcriptional and metabolic pathways can be dysregulated during acute and chronic MS lesion activity depriving OLs from critical acetyl-CoA biochemical mechanisms governing myelin lipid biosynthesis and at the same time altering the generation of iron metabolism that may drive ferroptotic mechanisms, leading to advancing neurodegeneration.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| |
Collapse
|
304
|
Seo H, Yoon Y, Kim S, Ghorbanian F, Tajdozian H, Jo S, Barman I, Lee S, Lee Y, Rahim MA, Hossain MS, Lee S, Song HY. Anti-tuberculosis effect of microbiome therapeutic PMC205 in extensively drug-resistant pulmonary tuberculosis in vivo. Int J Antimicrob Agents 2024; 64:107274. [PMID: 39002701 DOI: 10.1016/j.ijantimicag.2024.107274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis, and the increase in antibiotic resistance threatens humankind. Therefore, there is an urgent need to develop new anti-tuberculosis drugs that can overcome the limitations of existing drugs. Here, we report the anti-tuberculosis effect of microbiome therapeutic PMC205, a strain of Bacillus subtilis. METHODS The anti-tuberculosis activity of probiotics was evaluated in mouse models of lethal and latent pulmonary tuberculosis induced by high or low-dose infection of the extensively drug-resistant strain. Probiotics were administered by inhalation, and the burden of M. tuberculosis in the lungs, along with mortality and clinical observations, were monitored for 12 weeks and 8 months, respectively. For an in-depth understanding, analysis of the microbiome and inflammatory profile of the lung microenvironment and induction of autophagy in vitro were explored. RESULTS After inhalation administration of PMC205 for 3 months, the survival rate was 100%, unlike all deaths in the saline-treated group, and the burden of M. tuberculosis in the lungs was reduced by log 1.3 in the 8-month latent tuberculosis model. Moreover, PMC205 induced recovery of disrupted lung microflora, increased butyric acid, and suppressed excessive inflammation. It also promoted autophagy. CONCLUSIONS These results confirm PMC205's anti-tuberculosis effect, suggesting that it can be developed as an adjuvant to current antibiotic therapy to solve the drug-resistant tuberculosis problem.
Collapse
Affiliation(s)
- Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Hanieh Tajdozian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sujin Jo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Soyeon Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Saebim Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea.
| |
Collapse
|
305
|
Anurag Anand A, Amod A, Anwar S, Sahoo AK, Sethi G, Samanta SK. A comprehensive guide on screening and selection of a suitable AMP against biofilm-forming bacteria. Crit Rev Microbiol 2024; 50:859-878. [PMID: 38102871 DOI: 10.1080/1040841x.2023.2293019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Lately, antimicrobial resistance (AMR) is increasing at an exponential rate making it important to search alternatives to antibiotics in order to combat multi-drug resistant (MDR) bacterial infections. Out of the several antibacterial and antibiofilm strategies being tested, antimicrobial peptides (AMPs) have shown to give better hopes in terms of a long-lasting solution to the problem. To select a desired AMP, it is important to make right use of available tools and databases that aid in identification, classification, and analysis of the physiochemical properties of AMPs. To identify the targets of these AMPs, it becomes crucial to understand their mode-of-action. AMPs can also be used in combination with other antibacterial and antibiofilm agents so as to achieve enhanced efficacy against bacteria and their biofilms. Due to concerns regarding toxicity, stability, and bioavailability, strategizing drug formulation at an early-stage becomes crucial. Although there are few concerns regarding development of bacterial resistance to AMPs, the evolution of resistance to AMPs occurs extremely slowly. This comprehensive review gives a deep insight into the selection of the right AMP, deciding the right target and combination strategy along with the type of formulation needed, and the possible resistance that bacteria can develop to these AMPs.
Collapse
Affiliation(s)
- Ananya Anurag Anand
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Ayush Amod
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Sarfraz Anwar
- Department of Bioinformatics, University of Allahabad, Prayagraj, India
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sintu Kumar Samanta
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| |
Collapse
|
306
|
Leandro LF, Moraes TS, Damasceno JL, Veneziani RCS, Ambrosio SR, Bastos JK, Santiago MB, Pedroso RS, Martins CHG. Antibacterial, antibiofilm, and antivirulence potential of the main diterpenes from Copaifera spp. oleoresins against multidrug-resistant bacteria. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6975-6987. [PMID: 38619589 DOI: 10.1007/s00210-024-03077-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/30/2024] [Indexed: 04/16/2024]
Abstract
To evaluate the antibacterial, antibiofilm and antivirulence potential of the main diterpenes from Copaifera spp. oleoresins against multidrug-resistant (MDR) bacteria. Antimicrobial assays included determination of the Minimum Inhibitory Concentration (MIC), Minimum Bactericidal Concentration (MBC), Minimum Inhibitory Concentration of Biofilm (MICB50), as well as synergistic and antivirulence assays for eight diterpenes against MDR. The tests revealed that two diterpenes (named 1 and 5) showed the best results, with MIC and MBC between 12.5 and 50 μg/mL against most MDR bacteria. These diterpenes exhibited promising MICB50 in concentration between 3.12-25 μg/mL but showed no synergistic antimicrobial activity. In the assessment of antivirulence activity, diterpenes 1 and 5 inhibited only one of the virulence factors evaluated (Dnase) produced by some strains of S. aureus at subinhibitory concentration (6.25 μg/mL). Results obtained indicated that diterpenes isolated from Copaifera oleoresin plays an important part in the search of new antibacterial and antibiofilm agents that can act against MDR bacteria.
Collapse
Affiliation(s)
- L F Leandro
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, 38405-320, Brazil
| | - T S Moraes
- Nucleus of Research in Sciences and Technology, University of Franca - UNIFRAN, Franca, São Paulo, Brazil
| | - J L Damasceno
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, 38405-320, Brazil
| | - R C S Veneziani
- Nucleus of Research in Sciences and Technology, University of Franca - UNIFRAN, Franca, São Paulo, Brazil
| | - S R Ambrosio
- Nucleus of Research in Sciences and Technology, University of Franca - UNIFRAN, Franca, São Paulo, Brazil
| | - J K Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - M B Santiago
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, 38405-320, Brazil
| | - R S Pedroso
- Technical School of Health, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil
| | - C H G Martins
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, 38405-320, Brazil.
| |
Collapse
|
307
|
Kendell-Wall R, Nguyen JT, Salleras F, Kamboj AS, Diwen Tan SA, Manish Trivedi V, de Mello-Neto JM, Rodrigues Amaral R. Antimicrobial efficacy of Odontopaste in endodontics: a systematic review. Evid Based Dent 2024; 25:166. [PMID: 38538855 PMCID: PMC11436364 DOI: 10.1038/s41432-024-01000-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 09/29/2024]
Abstract
AIM To evaluate the efficacy of Odontopaste in reducing the microbial load in endodontics compared to other intracanal medicaments. MATERIALS AND METHODS The literature was electronically searched on PubMed, Google Scholar, Scopus, Ovid Medline and Web of Science. In-vitro, ex-vivo and in-vivo studies that evaluated the antimicrobial efficacy of Odontopaste were included. The risk of bias was assessed using the Quality Assessment Tool for In Vitro Studies. RESULTS A total of four in-vitro studies were included in the systematic review. One study showed that Odontopaste had significantly more microbial cell growth on roots in all dentine depths compared to other medicaments or test agents. Another study found that Odontopaste significantly decreased colony-forming units compared to propolis and chlorhexidine. Further results showed that Odontopaste did not significantly decrease microbial numbers when used in isolation. Additionally, combining Odontopaste and calcium hydroxide did not enhance the effectiveness of calcium hydroxide. The studies had a medium to high risk of bias. CONCLUSION There is insufficient high-quality evidence to assess the antimicrobial efficacy of Odontopaste compared to other intracanal medicaments. Further research is required to determine Odontopaste's efficacy as an antimicrobial medicament in endodontics.
Collapse
Affiliation(s)
- Rachael Kendell-Wall
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | - Jennifer-Thuy Nguyen
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | - Fauve Salleras
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | - Amandeep Singh Kamboj
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | - Serene Aimee Diwen Tan
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | - Vaidehi Manish Trivedi
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia
| | | | - Rodrigo Rodrigues Amaral
- College of Medicine and Dentistry, James Cook University, Campus Smithfield, Cairns, QLD, Australia.
| |
Collapse
|
308
|
Allend SO, de Oliveira Garcia M, da Cunha KF, Albernaz DTF, Panagio LA, Nakazato G, Reis GF, Oliveira TL, SeixasNeto ACP, Hartwig DD. Antibiofilm effect of biogenic silver nanoparticle alone and combined with polymyxin B against carbapenem-resistant Acinetobacter baumannii. Braz J Microbiol 2024; 55:2789-2796. [PMID: 39023813 PMCID: PMC11405624 DOI: 10.1007/s42770-024-01438-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Acinetobacter baumannii is a bacteria associated with nosocomial infections and outbreaks, difficult to control due to its antibiotic resistance, ability to survive in adverse conditions, and biofilm formation adhering to biotic and abiotic surfaces. Therefore, this study aimed to evaluate the antibiofilm activity of biogenic silver nanoparticle (Bio-AgNP) and polymyxin B alone and combined in biofilms formed by isolates of carbapenem-resistant A. baumannii (CR-Ab). In the biofilm formation inhibition assay, CR-Ab strains were exposed to different concentrations of the treatments before inducing biofilm formation, to determine the ability to inhibit/prevent bacterial biofilm formation. While in the biofilm rupture assay, the bacterial biofilm formation step was previously carried out and the adhered cells were exposed to different concentrations of the treatments to evaluate their ability to destroy the bacterial biofilm formed. All CR-Ab isolates and ATCC® 19606™ used in this study are strong biofilm formers. The antibiofilm activity of Bio-AgNP and polymyxin B against CR-Ab and ATCC® 19606™ demonstrated inhibitory and biofilm-disrupting activity. When used in combination, Bio-AgNP and polymyxin B inhibited 4.9-100% of biofilm formation in the CR-Ab isolates and ATCC® 19606™. Meanwhile, when Bio-AgNP and polymyxin B were combined, disruption of 6.8-77.8% of biofilm formed was observed. Thus, antibiofilm activity against CR-Ab was demonstrated when Bio-AgNP was used alone or in combination with polymyxin B, emerging as an alternative in the control of CR-Ab strains.
Collapse
Affiliation(s)
- Suzane Olachea Allend
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | - Marcelle de Oliveira Garcia
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | - Kamila Furtado da Cunha
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | - Déborah Trota Farias Albernaz
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | | | - Gerson Nakazato
- Department of Microbiology, State University of Londrina, Londrina, PR, CEP 86057-970, Brazil
| | - Guilherme Fonseca Reis
- Department of Microbiology, State University of Londrina, Londrina, PR, CEP 86057-970, Brazil
| | - Thaís Larré Oliveira
- Biotechnology Nucleus, Technological Development Center, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | - Amilton Clair Pinto SeixasNeto
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil
| | - Daiane Drawanz Hartwig
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
309
|
Adeyanju MM, Ademakinwa AN. Tyrosinase from the pulps of local cultivars of Musa spp: Purification, characterization, immobilization, and application in the batch production of l-3,4-dihydroxyphenylalanine. Prep Biochem Biotechnol 2024; 54:1098-1105. [PMID: 38445841 DOI: 10.1080/10826068.2024.2324084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Tyrosinase, an enzyme involved in browning reactions in plants/crops exposed to mechanical injury, was isolated from the pulp of some different locally available bananas (M. cavendish, M. acuminata, and M. paradisiaca). Tyrosinase from the pulps was extracted, purified, immobilized, and characterized. Thereafter, the potentials of the immobilized tyrosinase in the possible production of l-3,4-dihydroxyphenylalanine (L-DOPA) in an improvised batch reactor was exploited using tyrosine and ascorbate as the substrates. L-DOPA production was monitored via thin-layer chromatography and spectrophotometry (Arnow's method). L-DOPA is a drug that is used in the treatment of Parkinson's disease. Hence, this study exploited a non-chemical route for its synthesis using the tyrosinase obtained from the banana pulps. The purified tyrosinase had an optimum pH and temperature of 6.5 and 7.0, respectively. The molecular weight of the purified tyrosinase was 45 kDa. Quercetin and resorcinol both competitively inhibited the purified tyrosinase from the three cultivars. Immobilized M. cavendish tyrosinase produced the highest concentration (0.60 mM) of L-DOPA after 8 h in an improvised batch reactor. The tyrosinase in the banana pulps serves as a cheap and readily available green route for the possible production of L-DOPA.
Collapse
|
310
|
Peng B, Li H, Peng X. Understanding metabolic resistance strategy of clinically isolated antibiotic-resistant bacteria by proteomic approach. Expert Rev Proteomics 2024; 21:377-386. [PMID: 39387182 DOI: 10.1080/14789450.2024.2413439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Understanding the metabolic regulatory mechanisms leading to antibacterial resistance is important to develop effective control measures. AREAS COVERED In this review, we summarize the progress on metabolic mechanisms of antibiotic resistance in clinically isolated bacteria, as revealed using proteomic approaches. EXPERT OPINION Proteomic approaches are effective tools for uncovering clinically significant bacterial metabolic responses to antibiotics. Proteomics can disclose the associations between metabolic proteins, pathways, and networks with antibiotic resistance, and help identify their functional impact. The mechanisms by which metabolic proteins control the four generally recognized resistance mechanisms (decreased influx and targets, and increased efflux and enzymatic degradation) are particularly important. The proposed mechanism of reprogramming proteomics via key metabolites to enhance the killing efficiency of existing antibiotics needs attention.
Collapse
Affiliation(s)
- Bo Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, Guangzhou, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, Guangzhou, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xuanxian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, Guangzhou, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
311
|
Biswas R, Jangra B, Ashok G, Ravichandiran V, Mohan U. Current Strategies for Combating Biofilm-Forming Pathogens in Clinical Healthcare-Associated Infections. Indian J Microbiol 2024; 64:781-796. [PMID: 39282194 PMCID: PMC11399387 DOI: 10.1007/s12088-024-01221-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 02/07/2024] [Indexed: 09/18/2024] Open
Abstract
The biofilm formation by various pathogens causes chronic infections and poses severe threats to industry, healthcare, and society. They can form biofilm on surfaces of medical implants, heart valves, pacemakers, contact lenses, vascular grafts, urinary catheters, dialysis catheters, etc. These biofilms play a central role in bacterial persistence and antibiotic tolerance. Biofilm formation occurs in a series of steps, and any interference in these steps can prevent its formation. Therefore, the hunt to explore and develop effective anti-biofilm strategies became necessary to decrease the rate of biofilm-related infections. In this review, we highlighted and discussed the current therapeutic approaches to eradicate biofilm formation and combat drug resistance by anti-biofilm drugs, phytocompounds, antimicrobial peptides (AMPs), antimicrobial lipids (AMLs), matrix-degrading enzymes, nanoparticles, phagebiotics, surface coatings, photodynamic therapy (PDT), riboswitches, vaccines, and antibodies. The clinical validation of these findings will provide novel preventive and therapeutic strategies for biofilm-associated infections to the medical world.
Collapse
Affiliation(s)
- Rashmita Biswas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Bhawana Jangra
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab India
| | - Ganapathy Ashok
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Velayutham Ravichandiran
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| |
Collapse
|
312
|
Okaiyeto K, Gigliobianco MR, Di Martino P. Biogenic Zinc Oxide Nanoparticles as a Promising Antibacterial Agent: Synthesis and Characterization. Int J Mol Sci 2024; 25:9500. [PMID: 39273447 PMCID: PMC11395547 DOI: 10.3390/ijms25179500] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Nanotechnology has gained popularity in recent years due to its wide-ranging applications within the scientific community. The three main methods for synthesizing nanoparticles are physical, chemical, and biological. However, the adverse effects associated with physical and chemical methods have led to a growing interest in biological methods. Interestingly, green synthesis using plants has gained prominence in developing new treatments for bacterial infections. Zinc oxide nanoparticles (ZnO NPs) produced using environmentally friendly methods are more biocompatible and have potential applications as antibacterial agents in the biomedical field. As a result, this review discusses the green synthesis of ZnO NPs, factors influencing optimal synthesis, characterization techniques, and the antibacterial activity of some plant-mediated ZnO NPs. It also provides a comprehensive and analytical exploration of ZnO NP biosynthesis, the role of phytochemical compounds as reducing and stabilizing agents, the mechanism of action of their antibacterial properties and further highlights the challenges and prospects in this innovative research area.
Collapse
Affiliation(s)
- Kunle Okaiyeto
- Department of Pharmacy, University of "G. d'Annunzio" of Chieti and Pescara, Via dei Vestini, 1, 66100 Chieti, Italy
| | - Maria Rosa Gigliobianco
- Department of Pharmacy, University of "G. d'Annunzio" of Chieti and Pescara, Via dei Vestini, 1, 66100 Chieti, Italy
| | - Piera Di Martino
- Department of Pharmacy, University of "G. d'Annunzio" of Chieti and Pescara, Via dei Vestini, 1, 66100 Chieti, Italy
| |
Collapse
|
313
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
314
|
Cheng CK, Yi M, Wang L, Huang Y. Role of gasdermin D in inflammatory diseases: from mechanism to therapeutics. Front Immunol 2024; 15:1456244. [PMID: 39253076 PMCID: PMC11381298 DOI: 10.3389/fimmu.2024.1456244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory diseases compromise a clinically common and diverse group of conditions, causing detrimental effects on body functions. Gasdermins (GSDM) are pore-forming proteins, playing pivotal roles in modulating inflammation. Belonging to the GSDM family, gasdermin D (GSDMD) actively mediates the pathogenesis of inflammatory diseases by mechanistically regulating different forms of cell death, particularly pyroptosis, and cytokine release, in an inflammasome-dependent manner. Aberrant activation of GSDMD in different types of cells, such as immune cells, cardiovascular cells, pancreatic cells and hepatocytes, critically contributes to the persistent inflammation in different tissues and organs. The contributory role of GSDMD has been implicated in diabetes mellitus, liver diseases, cardiovascular diseases, neurodegenerative diseases, and inflammatory bowel disease (IBD). Clinically, alterations in GSDMD levels are potentially indicative to the occurrence and severity of diseases. GSDMD inhibition might represent an attractive therapeutic direction to counteract the progression of inflammatory diseases, whereas a number of GSDMD inhibitors have been shown to restrain GSDMD-mediated pyroptosis through different mechanisms. This review discusses the current understanding and future perspectives on the role of GSDMD in the development of inflammatory diseases, as well as the clinical insights of GSDMD alterations, and therapeutic potential of GSDMD inhibitors against inflammatory diseases. Further investigation on the comprehensive role of GSDM shall deepen our understanding towards inflammation, opening up more diagnostic and therapeutic opportunities against inflammatory diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Min Yi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
315
|
Machingauta A, Mukanganyama S. Antibacterial Activity and Proposed Mode of Action of Extracts from Selected Zimbabwean Medicinal Plants against Acinetobacter baumannii. Adv Pharmacol Pharm Sci 2024; 2024:8858665. [PMID: 39220823 PMCID: PMC11364482 DOI: 10.1155/2024/8858665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 06/27/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Acinetobacter baumannii was identified by the WHO as a priority pathogen in which the research and development of new antibiotics is urgently needed. Plant phytochemicals have potential as sources of new antimicrobials. The objective of the study was to determine the antibacterial activity of extracts of selected Zimbabwean medicinal plants against A. baumannii and determine their possible mode of action. Extracts were prepared from the leaves of the eight plants including the bark of Erythrina abyssinica using solvents of different polarities. Antibacterial activity was evaluated using the microbroth dilution method coupled with the in vitro iodonitrotetrazolium colorimetric assay. The effect of the extracts on membrane integrity was determined by quantifying the amount of protein and nucleic acid leaked from the cells after exposure to the extracts. The effects of the extracts on biofilms were investigated. Toxicity studies were carried out using sheep erythrocytes and murine peritoneal cells. Seven out of eight evaluated plant extracts were found to have antibacterial activity. The Combretum apiculatum acetonie (CAA) extract showed the highest inhibitory activity against A. baumannii with a minimal inhibitory concentration of 125 µg/mL. The minimum inhibitory concentration (MIC) of the CAA extract caused a protein leakage of 32 µg/mL from A. baumannii. The Combretum apiculatum acetonie (CAA), C. apiculatum methanolic (CAM), Combretum zeyheri methanolic (CZM), and Erythrina abyssinica methanolic (EAM) extracts inhibited A. baumannii biofilm formation. The EAM extract was shown to disrupt mature biofilms. The potent extracts were nontoxic to sheep erythrocytes and mouse peritoneal cells. The activities shown by the extracts indicate that the plants have potential as sources of effective antibacterial and antibiofilm formation agents against A. baumannii.
Collapse
Affiliation(s)
- Auxillia Machingauta
- Bio-Molecular Interactions Analyses GroupDepartment of Biotechnology and BiochemistryUniversity of Zimbabwe, Mt Pleasant, P.O. Box 167, Harare, Zimbabwe
| | - Stanley Mukanganyama
- Department of TherapeuticsNatural Products Research UnitAfrican Institute of Biomedical Science and TechnologyWilkins Hospital, Block C, Corner J. Tongogara and R. Tangwena, Harare, Zimbabwe
| |
Collapse
|
316
|
Mishra A, Tabassum N, Aggarwal A, Kim YM, Khan F. Artificial Intelligence-Driven Analysis of Antimicrobial-Resistant and Biofilm-Forming Pathogens on Biotic and Abiotic Surfaces. Antibiotics (Basel) 2024; 13:788. [PMID: 39200087 PMCID: PMC11351874 DOI: 10.3390/antibiotics13080788] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
The growing threat of antimicrobial-resistant (AMR) pathogens to human health worldwide emphasizes the need for more effective infection control strategies. Bacterial and fungal biofilms pose a major challenge in treating AMR pathogen infections. Biofilms are formed by pathogenic microbes encased in extracellular polymeric substances to confer protection from antimicrobials and the host immune system. Biofilms also promote the growth of antibiotic-resistant mutants and latent persister cells and thus complicate therapeutic approaches. Biofilms are ubiquitous and cause serious health risks due to their ability to colonize various surfaces, including human tissues, medical devices, and food-processing equipment. Detection and characterization of biofilms are crucial for prompt intervention and infection control. To this end, traditional approaches are often effective, yet they fail to identify the microbial species inside biofilms. Recent advances in artificial intelligence (AI) have provided new avenues to improve biofilm identification. Machine-learning algorithms and image-processing techniques have shown promise for the accurate and efficient detection of biofilm-forming microorganisms on biotic and abiotic surfaces. These advancements have the potential to transform biofilm research and clinical practice by allowing faster diagnosis and more tailored therapy. This comprehensive review focuses on the application of AI techniques for the identification of biofilm-forming pathogens in various industries, including healthcare, food safety, and agriculture. The review discusses the existing approaches, challenges, and potential applications of AI in biofilm research, with a particular focus on the role of AI in improving diagnostic capacities and guiding preventative actions. The synthesis of the current knowledge and future directions, as described in this review, will guide future research and development efforts in combating biofilm-associated infections.
Collapse
Affiliation(s)
- Akanksha Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Ashish Aggarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
317
|
Motta EVS, de Jong TK, Gage A, Edwards JA, Moran NA. Glyphosate effects on growth and biofilm formation in bee gut symbionts and diverse associated bacteria. Appl Environ Microbiol 2024; 90:e0051524. [PMID: 39012136 PMCID: PMC11337805 DOI: 10.1128/aem.00515-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Biofilm formation is a common adaptation enabling bacteria to thrive in various environments and withstand external pressures. In the context of host-microbe interactions, biofilms play vital roles in establishing microbiomes associated with animals and plants and are used by opportunistic microbes to facilitate survival within hosts. Investigating biofilm dynamics, composition, and responses to environmental stressors is crucial for understanding microbial community assembly and biofilm regulation in health and disease. In this study, we explore in vivo colonization and in vitro biofilm formation abilities of core members of the honey bee (Apis mellifera) gut microbiota. Additionally, we assess the impact of glyphosate, a widely used herbicide with antimicrobial properties, and a glyphosate-based herbicide formulation on growth and biofilm formation in bee gut symbionts as well as in other biofilm-forming bacteria associated with diverse animals and plants. Our results demonstrate that several strains of core bee gut bacterial species can colonize the bee gut, which probably depends on their ability to form biofilms. Furthermore, glyphosate exposure elicits variable effects on bacterial growth and biofilm formation. In some instances, the effects correlate with the bacteria's ability to encode a susceptible or tolerant version of the enzyme inhibited by glyphosate in the shikimate pathway. However, in other instances, no such correlation is observed. Testing the herbicide formulation further complicates comparisons, as results often diverge from glyphosate exposure alone, suggesting that co-formulants influence bacterial growth and biofilm formation. These findings highlight the nuanced impacts of environmental stressors on microbial biofilms, with both ecological and host health-related implications. IMPORTANCE Biofilms are essential for microbial communities to establish and thrive in diverse environments. In the honey bee gut, the core microbiota member Snodgrassella alvi forms biofilms, potentially aiding the establishment of other members and promoting interactions with the host. In this study, we show that specific strains of other core members, including Bifidobacterium, Bombilactobacillus, Gilliamella, and Lactobacillus, also form biofilms in vitro. We then examine the impact of glyphosate, a widely used herbicide that can disrupt the bee microbiota, on bacterial growth and biofilm formation. Our findings demonstrate the diverse effects of glyphosate on biofilm formation, ranging from inhibition to enhancement, reflecting observations in other beneficial or pathogenic bacteria associated with animals and plants. Thus, glyphosate exposure may influence bacterial growth and biofilm formation, potentially shaping microbial establishment on host surfaces and impacting health outcomes.
Collapse
Affiliation(s)
- Erick V. S. Motta
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Entomology, Texas A&M University, College Station, Texas, USA
| | - Tyler K. de Jong
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Alejandra Gage
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Joseph A. Edwards
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, Texas, USA
| | - Nancy A. Moran
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
318
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: a review. Front Pharmacol 2024; 15:1444781. [PMID: 39221153 PMCID: PMC11362070 DOI: 10.3389/fphar.2024.1444781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Antibacterial drug resistance poses a significant challenge to modern healthcare systems, threatening our ability to effectively treat bacterial infections. This review aims to provide a comprehensive overview of the types and mechanisms of antibacterial drug resistance. To achieve this aim, a thorough literature search was conducted to identify key studies and reviews on antibacterial resistance mechanisms, strategies and next-generation antimicrobials to contain antimicrobial resistance. In this review, types of resistance and major mechanisms of antibacterial resistance with examples including target site modifications, decreased influx, increased efflux pumps, and enzymatic inactivation of antibacterials has been discussed. Moreover, biofilm formation, and horizontal gene transfer methods has also been included. Furthermore, measures (interventions) taken to control antimicrobial resistance and next-generation antimicrobials have been discussed in detail. Overall, this review provides valuable insights into the diverse mechanisms employed by bacteria to resist the effects of antibacterial drugs, with the aim of informing future research and guiding antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of clinical pharmacy, College of medicine and health sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of environmental health science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
319
|
Li JX, Qu YD, Xia CL, Zhang W, Wang SS, Ou SJ, Yang Y, Qi Y, Xu CP. Analysis of PANoptosis-related ceRNA network reveals lncRNA MIR17HG involved in osteogenic differentiation inhibition impaired by tumor necrosis factor-α. Mol Biol Rep 2024; 51:909. [PMID: 39145884 PMCID: PMC11327206 DOI: 10.1007/s11033-024-09810-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/19/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Inflammatory cytokines such as Interleukin 1β(IL1β), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.
Collapse
Affiliation(s)
- Jia-Xuan Li
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu-Dun Qu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Chang-Liang Xia
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Wei Zhang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Song-Song Wang
- School of Medicine, XiaMen University, Xiamen, Fujian, China
| | - Shuan-Ji Ou
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yang Yang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yong Qi
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| | - Chang-Peng Xu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
320
|
Lisjak D, Alić I, Šimunić I, Mitrečić D. Transplantation of neural stem cells improves recovery of stroke-affected mice and induces cell-specific changes in GSDMD and MLKL expression. Front Mol Neurosci 2024; 17:1439994. [PMID: 39210936 PMCID: PMC11358122 DOI: 10.3389/fnmol.2024.1439994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Stroke, the second leading cause of death and disability in Europe, is primarily caused by interrupted blood supply, leading to ischemia-reperfusion (IR) injury and subsequent neuronal death. Current treatment options are limited, highlighting the need for novel therapies. Neural stem cells (NSCs) have shown promise in treating various neurological disorders, including stroke. However, the underlying mechanisms of NSC-mediated recovery remain unclear. Methods Eighty C57Bl/6-Tyrc-Brd mice underwent ischemic stroke induction and were divided into four groups: sham, stroke-affected, stroke-affected with basal cell medium injection, and stroke-affected with NSCs transplantation. NSCs, isolated from mouse embryos, were stereotaxically transplanted into the stroke-affected brains. Magnetic resonance imaging (MRI) and neurological scoring were used to assess recovery. Immunohistochemical analysis and gene expression assays were performed to evaluate pyroptosis and necroptosis markers. Results NSC transplantation significantly improved neurological recovery compared to control groups. In addition, although not statistically significant, NSCs reduced stroke volume. Immunohistochemical analysis revealed upregulation of Gasdermin D (GSDMD) expression post-stroke, predominantly in microglia and astrocytes. However, NSC transplantation led to a reduction in GSDMD signal intensity in astrocytes, suggesting an effect of NSCs on GSDMD activity. Furthermore, NSCs downregulated Mixed Lineage Kinase Domain-Like Protein (Mlkl) expression, indicating a reduction in necroptosis. Immunohistochemistry demonstrated decreased phosphorylated MLKL (pMLKL) signal intensity in neurons while stayed the same in astrocytes following NSC transplantation, along with increased distribution in microglia. Discussion NSC transplantation holds therapeutic potential in stroke recovery by targeting pyroptosis and necroptosis pathways. These findings shed light on the mechanisms underlying NSC-mediated neuroprotection and support their further exploration as a promising therapy for stroke patients.
Collapse
Affiliation(s)
- Damir Lisjak
- Laboratory for Stem Cells, Department for Regenerative Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Alić
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Iva Šimunić
- Laboratory for Stem Cells, Department for Regenerative Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Department for Regenerative Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
321
|
Demontier E, Ster C, Chamberland S, Ramanathan S, Dufour S, Malouin F. Biofilm Dairy Foods Review: Effect of biofilm production on antimicrobial susceptibility of Staphylococcus aureus bovine mastitis strains from the most prevalent Canadian spa types. J Dairy Sci 2024:S0022-0302(24)01061-0. [PMID: 39122151 DOI: 10.3168/jds.2024-25238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Staphylococcus aureus intramammary infections often leads to clinical and subclinical mastitis in dairy cattle. Prediction of disease evolution and treatment efficacy based on the characteristics of disease-causing strains of S. aureus would significantly improve management of dairy herds. To study the impact of biofilm production and the influence of genetic lineage, we selected S. aureus isolates from the most prevalent Canadian spa types associated with bovine mastitis. Antimicrobial susceptibility in planktonic growth and for bacteria embedded in biofilm was compared. PCR was used to detect the bap gene responsible for atypical biofilm formation. All Canadian spa types from dairy cattle were susceptible to the 8 antimicrobial agents tested. Only strain sa3493 from spa type t267 showed a resistance to pirlimycin. However, bacteria producing larger amounts of biofilms better survived the bactericidal action of antimicrobial agents even when exposed to concentrations 64 folds higher than the minimal inhibitory concentration determined for planktonic cultures. Pirlimycin was more effective on bacteria producing low to moderate levels of biofilm compared with vancomycin or ceftiofur. Antimicrobial agents did not affect the viability of spa types t13401 and t605 that were high biofilm producers. While both these spa types produced high amounts of biofilm, only t605 possessed the bap gene. We also found a close relationship between DIM at sampling and the presence of spa type t605 isolates. These results suggest that detection of S. aureus spa type may help predict the effectiveness of antimicrobial therapy and that some spa types are more likely to be retrieved toward the end of the lactation.
Collapse
Affiliation(s)
- E Demontier
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1K 2R1; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2
| | - C Ster
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1K 2R1; New address: Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2
| | - S Chamberland
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1K 2R1; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2
| | - S Ramanathan
- Département d'immunologie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada, J1H 5N4; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2
| | - S Dufour
- Département de pathologie et de microbiologie, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada, J2S 2M2; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2
| | - F Malouin
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1K 2R1; Regroupement stratégique FRQNT pour un lait de qualité optimale, Op+lait, Université de Montréal, St-Hyacinthe, QC, Canada, J2S 2M2.
| |
Collapse
|
322
|
Zallocchi M, Vijayakumar S, Fleegel J, Batalkina L, Brunette KE, Shukal D, Chen Z, Devuyst O, Liu H, He DZZ, Imami AS, Hamoud ARA, McCullumsmith R, Conda-Sheridan M, De Campos LJ, Zuo J. Piplartine attenuates aminoglycoside-induced TRPV1 activity and protects from hearing loss in mice. Sci Transl Med 2024; 16:eadn2140. [PMID: 39110778 PMCID: PMC11392653 DOI: 10.1126/scitranslmed.adn2140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/17/2024] [Accepted: 06/28/2024] [Indexed: 09/15/2024]
Abstract
Hearing loss is a major health concern in our society, affecting more than 400 million people worldwide. Among the causes, aminoglycoside therapy can result in permanent hearing loss in 40% to 60% of patients receiving treatment, and despite these high numbers, no drug for preventing or treating this type of hearing loss has yet been approved by the US Food and Drug Administration. We have previously conducted high-throughput screenings of bioactive compounds, using zebrafish as our discovery platform, and identified piplartine as a potential therapeutic molecule. In the present study, we expanded this work and characterized piplartine's physicochemical and therapeutic properties. We showed that piplartine had a wide therapeutic window and neither induced nephrotoxicity in vivo in zebrafish nor interfered with aminoglycoside antibacterial activity. In addition, a fluorescence-based assay demonstrated that piplartine did not inhibit cytochrome C activity in microsomes. Coadministration of piplartine protected from kanamycin-induced hair cell loss in zebrafish and protected hearing function, outer hair cells, and presynaptic ribbons in a mouse model of kanamycin ototoxicity. Last, we investigated piplartine's mechanism of action by phospho-omics, immunoblotting, immunohistochemistry, and molecular dynamics experiments. We found an up-regulation of AKT1 signaling in the cochleas of mice cotreated with piplartine. Piplartine treatment normalized kanamycin-induced up-regulation of TRPV1 expression and modulated the gating properties of this receptor. Because aminoglycoside entrance to the inner ear is, in part, mediated by TRPV1, these results suggested that by regulating TRPV1 expression, piplartine blocked aminoglycoside's entrance, thereby preventing the long-term deleterious effects of aminoglycoside accumulation in the inner ear compartment.
Collapse
Affiliation(s)
| | | | | | | | | | - Dhaval Shukal
- Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Zhiyong Chen
- Institute of Physiology, University of Zürich, Zürich CH-8057, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, University of Zürich, Zürich CH-8057, Switzerland
| | - Huizhan Liu
- Creighton University School of Medicine, Omaha, NE 68178, USA
| | - David Z. Z. He
- Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Ali Sajid Imami
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | | | - Robert McCullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
- Neurosciences Institute, ProMedica, Toledo, OH 43614, USA
| | | | | | - Jian Zuo
- Creighton University School of Medicine, Omaha, NE 68178, USA
- Ting Therapeutics LLC, La Jolla, CA 92037, USA
| |
Collapse
|
323
|
Abdelhamid MAA, Khalifa HO, Yoon HJ, Ki MR, Pack SP. Microbial Immobilized Enzyme Biocatalysts for Multipollutant Mitigation: Harnessing Nature's Toolkit for Environmental Sustainability. Int J Mol Sci 2024; 25:8616. [PMID: 39201301 PMCID: PMC11355015 DOI: 10.3390/ijms25168616] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
The ever-increasing presence of micropollutants necessitates the development of environmentally friendly bioremediation strategies. Inspired by the remarkable versatility and potent catalytic activities of microbial enzymes, researchers are exploring their application as biocatalysts for innovative environmental cleanup solutions. Microbial enzymes offer remarkable substrate specificity, biodegradability, and the capacity to degrade a wide array of pollutants, positioning them as powerful tools for bioremediation. However, practical applications are often hindered by limitations in enzyme stability and reusability. Enzyme immobilization techniques have emerged as transformative strategies, enhancing enzyme stability and reusability by anchoring them onto inert or activated supports. These improvements lead to more efficient pollutant degradation and cost-effective bioremediation processes. This review delves into the diverse immobilization methods, showcasing their success in degrading various environmental pollutants, including pharmaceuticals, dyes, pesticides, microplastics, and industrial chemicals. By highlighting the transformative potential of microbial immobilized enzyme biocatalysts, this review underscores their significance in achieving a cleaner and more sustainable future through the mitigation of micropollutant contamination. Additionally, future research directions in areas such as enzyme engineering and machine learning hold immense promise for further broadening the capabilities and optimizing the applications of immobilized enzymes in environmental cleanup.
Collapse
Affiliation(s)
- Mohamed A. A. Abdelhamid
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
- Department of Botany and Microbiology, Faculty of Science, Minia University, Minia 61519, Egypt
- Faculty of Education and Art, Sohar University, Sohar 311, Oman
| | - Hazim O. Khalifa
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 1555, United Arab Emirates;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Hyo Jik Yoon
- Institute of Natural Science, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea;
| | - Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
- Institute of Industrial Technology, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea; (M.A.A.A.); (M.-R.K.)
| |
Collapse
|
324
|
Kunnath AP, Suodha Suoodh M, Chellappan DK, Chellian J, Palaniveloo K. Bacterial Persister Cells and Development of Antibiotic Resistance in Chronic Infections: An Update. Br J Biomed Sci 2024; 81:12958. [PMID: 39170669 PMCID: PMC11335562 DOI: 10.3389/bjbs.2024.12958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
The global issue of antimicrobial resistance poses significant challenges to public health. The World Health Organization (WHO) has highlighted it as a major global health threat, causing an estimated 700,000 deaths worldwide. Understanding the multifaceted nature of antibiotic resistance is crucial for developing effective strategies. Several physiological and biochemical mechanisms are involved in the development of antibiotic resistance. Bacterial cells may escape the bactericidal actions of the drugs by entering a physiologically dormant state known as bacterial persistence. Recent findings in this field suggest that bacterial persistence can be one of the main sources of chronic infections. The antibiotic tolerance developed by the persister cells could tolerate high levels of antibiotics and may give rise to persister offspring. These persister offspring could be attributed to antibiotic resistance mechanisms, especially in chronic infections. This review attempts to shed light on persister-induced antibiotic resistance and the current therapeutic strategies.
Collapse
Affiliation(s)
- Anil Philip Kunnath
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Mohamed Suodha Suoodh
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kishneth Palaniveloo
- Institute of Ocean and Earth Sciences, Institute for Advanced Studies Building, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
325
|
Oates WA, Anastasiou AD. A novel microfluidic tool for the evaluation of local drug delivery systems in simulated in vivo conditions. LAB ON A CHIP 2024; 24:3840-3849. [PMID: 39045628 DOI: 10.1039/d4lc00181h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
A 3D-printed microfluidic tool for assessing local drug delivery systems (LDD) in simulated in vivo conditions was developed and evaluated. The device was designed considering the oral environment and dental applications, and it was fabricated with a high-precision resin 3D printer. Chitosan scaffolds loaded with different concentrations of doxycycline were used for evaluating our device. The concentration of the released drug was measured through in-line UV-VIS spectroscopy, and to verify the repeatability and accuracy of our measurements, comparisons with standard HPLC results were made (5% deviation). Cumulative drug release profiles in steady-state conditions were obtained and compared to the Weibull model. The behaviour of the LDD system in a dynamic environment was also evaluated during experiments where step changes in pH were introduced. It was demonstrated that under infection-like conditions, there is an immediate response from the polymer and a clear increase in the concentration of the released drug. Continuous flow and recirculation experiments were also conducted, revealing significant differences in the drug release profiles. Specifically, in the case of continuous flow, the quantity of the released drug is much higher due to the higher driving force for diffusion (concentration gradient). Overall, the proposed microfluidic tool proved to be ideal for evaluating LDD systems, as the in vivo microenvironment can be replicated in a better way than with currently used standard systems.
Collapse
Affiliation(s)
- William A Oates
- Lab of Complex Fluids and Microfluidics, Department of Chemical Engineering, University of Manchester, Manchester, M1 9PL, UK.
| | - Antonios D Anastasiou
- Lab of Complex Fluids and Microfluidics, Department of Chemical Engineering, University of Manchester, Manchester, M1 9PL, UK.
| |
Collapse
|
326
|
Gross T, Ledernez LA, Birrer L, Bergmann ME, Altenburger MJ. Guided Plasma Application in Dentistry-An Alternative to Antibiotic Therapy. Antibiotics (Basel) 2024; 13:735. [PMID: 39200035 PMCID: PMC11350922 DOI: 10.3390/antibiotics13080735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Cold atmospheric plasma (CAP) is a promising alternative to antibiotics and chemical substances in dentistry that can reduce the risk of unwanted side effects and bacterial resistance. AmbiJet is a device that can ignite and deliver plasma directly to the site of action for maximum effectiveness. The aim of the study was to investigate its antimicrobial efficacy and the possible development of bacterial resistance. The antimicrobial effect of the plasma was tested under aerobic and anaerobic conditions on bacteria (five aerobic, three anaerobic (Gram +/-)) that are relevant in dentistry. The application times varied from 1 to 7 min. Possible bacterial resistance was evaluated by repeated plasma applications (10 times in 50 days). A possible increase in temperature was measured. Plasma effectively killed 106 seeded aerobic and anaerobic bacteria after an application time of 1 min per 10 mm2. Neither the development of resistance nor an increase in temperature above 40 °C was observed, so patient discomfort can be ruled out. The plasma treatment proved to be effective under anaerobic conditions, so the influence of ROS can be questioned. Our results show that AmbiJet efficiently eliminates pathogenic oral bacteria. Therefore, it can be advocated for clinical therapeutic use.
Collapse
Affiliation(s)
- Tara Gross
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center–University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (T.G.); (L.B.)
- Center for Tissue Replacement, Regeneration & Neogenesis (GERN), Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, 79108 Freiburg, Germany
| | - Loic Alain Ledernez
- Laboratory for Sensors, Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110 Freiburg, Germany; (L.A.L.); (M.E.B.)
| | - Laurent Birrer
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center–University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (T.G.); (L.B.)
- Center for Tissue Replacement, Regeneration & Neogenesis (GERN), Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, 79108 Freiburg, Germany
- Laboratory for Sensors, Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110 Freiburg, Germany; (L.A.L.); (M.E.B.)
| | - Michael Eckhard Bergmann
- Laboratory for Sensors, Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110 Freiburg, Germany; (L.A.L.); (M.E.B.)
| | - Markus Jörg Altenburger
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center–University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (T.G.); (L.B.)
- Center for Tissue Replacement, Regeneration & Neogenesis (GERN), Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, 79108 Freiburg, Germany
| |
Collapse
|
327
|
Al Sultan A, Rattray Z, Rattray NJW. Cytotoxicity and toxicoproteomics analysis of thiazolidinedione exposure in human-derived cardiomyocytes. J Appl Toxicol 2024; 44:1214-1235. [PMID: 38654465 DOI: 10.1002/jat.4613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/16/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Thiazolidinediones (TZDs) (e.g. pioglitazone and rosiglitazone), known insulin sensitiser agents for type II diabetes mellitus, exhibit controversial effects on cardiac tissue. Despite consensus on their association with increased heart failure risk, limiting TZD use in diabetes management, the underlying mechanisms remain uncharacterised. Herein, we report a comprehensive in vitro investigation utilising a novel toxicoproteomics pipeline coupled with cytotoxicity assays in human adult cardiomyocytes to elucidate mechanistic insights into TZD cardiotoxicity. The cytotoxicity assay findings showed a significant loss of mitochondrial adenosine triphosphate production upon exposure to either TZD agents, which may underpin TZD cardiotoxicity. Our toxicoproteomics analysis revealed that mitochondrial dysfunction primarily stems from oxidative phosphorylation impairment, with distinct signalling mechanisms observed for both agents. The type of cell death differed strikingly between the two agents, with rosiglitazone exhibiting features of caspase-dependent apoptosis and pioglitazone implicating mitochondrial-mediated necroptosis, as evidenced by the protein upregulation in the phosphoglycerate mutase family 5-dynamin-related protein 1 axis. Furthermore, our analysis revealed additional mechanistic aspects of cardiotoxicity, showcasing drug specificity. The downregulation of various proteins involved in protein machinery and protein processing in the endoplasmic reticulum was observed in rosiglitazone-treated cells, implicating proteostasis in the rosiglitazone cardiotoxicity. Regarding pioglitazone, the findings suggested the potential activation of the interplay between the complement and coagulation systems and the disruption of the cytoskeletal architecture, which was primarily mediated through the integrin-signalling pathways responsible for pioglitazone-induced myocardial contractile failure. Collectively, this study unlocks substantial mechanistic insight into TZD cardiotoxicity, providing the rationale for future optimisation of antidiabetic therapies.
Collapse
Affiliation(s)
- Abdullah Al Sultan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Faculty of Pharmacy, Kuwait University, Safat, Kuwait
| | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Nicholas J W Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
328
|
Mini M, Jayakumar D, Kumar P. In-silico and in-vitro assessment of the antibiofilm potential of azo dye, carmoisine against Pseudomonas aeruginosa. J Biomol Struct Dyn 2024; 42:6700-6710. [PMID: 37485898 DOI: 10.1080/07391102.2023.2237579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Biofilm is a community of microorganisms attached to the substrate and plays a significant role in microbial pathogenesis and medical device-related infection. Pseudomonas aeruginosa (PA) is a highly infectious gram-negative opportunistic biofilm-forming bacterium with high antibiotic resistance. Several reports underscore the antimicrobial activity of natural and synthetic food coloring agents, including carmoisine, turmeric dye, red amaranth dye, and phloxine B. However, their ability to suppress the PA biofilm is not clearly understood. Carmoisine is a red-colored synthetic azo dye containing naphthalene subunits and sulfonic groups and is widely used as a food coloring agent. This study investigated the antibiofilm potential and possible mechanism of biofilm inhibition by carmoisine against PA. Computational studies through molecular docking revealed that carmoisine strongly binds to QS regulator LasR (-12.7) and relatively less strongly but significantly with WspR (-6.9). Further analysis of the docked LasR-carmoisine complex using 100 ns MD simulation (Desmond, Schrödinger) validated the bonding strength and stability. Crystal violet assay, triphenyl tetrazolium chloride salt assay, and confocal microscopic studies were adopted for biofilm quantification, and the results indicated the dose-dependent antibiofilm activity of carmoisine against PA. We hypothesise that the carmoisine-mediated reduction of biofilm in PA is due to its interaction with LasR and interference with the QS system. The computational and biochemical analysis of another compound, 1,2-naphthoquinone-4-sulphonic acid, reiterated the role of the naphthalene ring in biofilm inhibition. Hence, this work will pave the way for the future discovery of antibiofilm drugs based on naphthalene ring-based lead compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Minsa Mini
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Devi Jayakumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Praveen Kumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
329
|
Dixit S, Varshney S, Gupta D, Sharma S. Factors affecting biofilm formation by bacteria on fabrics. Int Microbiol 2024; 27:1111-1123. [PMID: 38057457 DOI: 10.1007/s10123-023-00460-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023]
Abstract
Fabrics act as fomites for microorganisms, thereby playing a significant role in infection transmission, especially in the healthcare and hospitality sectors. This study aimed to examine the biofilm formation ability of four nosocomial infection-causing bacteria (Acinetobacter calcoaceticus, Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus) on cotton, polyester, polyester-cotton blend, silk, wool, viscose, and nylon, used frequently in the healthcare sector, by qualitative and quantitative methods. The impact of temperature, pH, and relative humidity (RH) on biofilm formation was also assessed. P. aeruginosa and S. aureus were strong biofilm producers, while E. coli produced weak biofilm. Wool (maximum roughness) showed the highest bacterial load, while silk (lowest roughness) showed the least. P. aeruginosa exhibited a higher load on all fabrics, than other test bacteria. Extracellular polymeric substances were characterized by infrared spectroscopy. Roughness of biofilms was assessed by atomic force microscopy. For biofilm formation, optimum temperature, pH, and RH were 30 °C, 7.0, and 62%, respectively. MgCl2 and CaCl2 were the most effective in removing bacterial biofilm. In conclusion, biofilm formation was observed to be influenced by the type of fabric, bacteria, and environmental conditions. Implementing recommended guidelines for the effective disinfection of fabrics is crucial to curb the risk of nosocomial infections. In addition, designing modified healthcare fabrics that inhibit pathogen load could be an effective method to mitigate the transmission of infections.
Collapse
Affiliation(s)
- Shweta Dixit
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Swati Varshney
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
- Present address: Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Deepti Gupta
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Shilpi Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
330
|
Haque PS, Kapur N, Barrett TA, Theiss AL. Mitochondrial function and gastrointestinal diseases. Nat Rev Gastroenterol Hepatol 2024; 21:537-555. [PMID: 38740978 PMCID: PMC12036329 DOI: 10.1038/s41575-024-00931-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Mitochondria are dynamic organelles that function in cellular energy metabolism, intracellular and extracellular signalling, cellular fate and stress responses. Mitochondria of the intestinal epithelium, the cellular interface between self and enteric microbiota, have emerged as crucial in intestinal health. Mitochondrial dysfunction occurs in gastrointestinal diseases, including inflammatory bowel diseases and colorectal cancer. In this Review, we provide an overview of the current understanding of intestinal epithelial cell mitochondrial metabolism, function and signalling to affect tissue homeostasis, including gut microbiota composition. We also discuss mitochondrial-targeted therapeutics for inflammatory bowel diseases and colorectal cancer and the evolving concept of mitochondrial impairment as a consequence versus initiator of the disease.
Collapse
Affiliation(s)
- Parsa S Haque
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Neeraj Kapur
- Department of Medicine, Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Terrence A Barrett
- Department of Medicine, Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY, USA
- Lexington Veterans Affairs Medical Center Kentucky, Lexington, KY, USA
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA.
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA.
| |
Collapse
|
331
|
Walsh D, Parmenter C, Bakker SE, Lithgow T, Traven A, Harrison F. A new model of endotracheal tube biofilm identifies combinations of matrix-degrading enzymes and antimicrobials able to eradicate biofilms of pathogens that cause ventilator-associated pneumonia. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001480. [PMID: 39088248 PMCID: PMC11541551 DOI: 10.1099/mic.0.001480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024]
Abstract
Ventilator-associated pneumonia is defined as pneumonia that develops in a patient who has been on mechanical ventilation for more than 48 hours through an endotracheal tube. It is caused by biofilm formation on the indwelling tube, which introduces pathogenic microbes such as Pseudomonas aeruginosa, Klebsiella pneumoniae and Candida albicans into the patient's lower airways. Currently, there is a lack of accurate in vitro models of ventilator-associated pneumonia development. This greatly limits our understanding of how the in-host environment alters pathogen physiology and the efficacy of ventilator-associated pneumonia prevention or treatment strategies. Here, we showcase a reproducible model that simulates the biofilm formation of these pathogens in a host-mimicking environment and demonstrate that the biofilm matrix produced differs from that observed in standard laboratory growth medium. In our model, pathogens are grown on endotracheal tube segments in the presence of a novel synthetic ventilated airway mucus medium that simulates the in-host environment. Matrix-degrading enzymes and cryo-scanning electron microscopy were employed to characterize the system in terms of biofilm matrix composition and structure, as compared to standard laboratory growth medium. As seen in patients, the biofilms of ventilator-associated pneumonia pathogens in our model either required very high concentrations of antimicrobials for eradication or could not be eradicated. However, combining matrix-degrading enzymes with antimicrobials greatly improved the biofilm eradication of all pathogens. Our in vitro endotracheal tube model informs on fundamental microbiology in the ventilator-associated pneumonia context and has broad applicability as a screening platform for antibiofilm measures including the use of matrix-degrading enzymes as antimicrobial adjuvants.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Chris Parmenter
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham, UK
| | | | - Trevor Lithgow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
332
|
Chakraborty S, Wei D, Tran M, Lang FF, Newman RA, Yang P. PBI-05204, a supercritical CO 2 extract of Nerium oleander, suppresses glioblastoma stem cells by inhibiting GRP78 and inducing programmed necroptotic cell death. Neoplasia 2024; 54:101008. [PMID: 38823209 PMCID: PMC11177059 DOI: 10.1016/j.neo.2024.101008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Successful treatment of glioblastoma multiforme (GBM), an aggressive form of primary brain neoplasm, mandates the need to develop new therapeutic strategies. In this study, we investigated the potential of PBI-05204 in targeting GBM stem cells (GSCs) and the underlying mechanisms. Treatment with PBI-05204 significantly reduced both the number and size of tumor spheres derived from patient-derived GSCs (GBM9, GSC28 and TS543), and suppressed the tumorigenesis of GBM9 xenografts. Moreover, PBI-05204 treatment led to a significant decrease in the expression of CD44 and NANOG, crucial markers of progenitor stem cells, in GBM9 and GSC28 GSCs. This treatment also down-regulated GRP78 expression in both GSC types. Knocking down GRP78 expression through GRP78 siRNA transfection in GBM9 and GSC28 GSCs also resulted in reduced spheroid size and CD44 expression. Combining PBI-05204 with GRP78 siRNA further decreased spheroid numbers compared to GRP78 siRNA treatment alone. PBI-05204 treatment led to increased expression of pRIP1K and pRIP3K, along with enhanced binding of RIPK1/RIPK3 in GBM9 and GSC28 cells, resembling the effects observed in GRP78-silenced GSCs, suggesting that PBI-05204 induced necroptosis in these cells. Furthermore, oleandrin, a principle active cardiac glycoside component of PBI-05204, showed the ability to inhibit the self-renewal capacity in GSCs. These findings highlight the potential of PBI-05204 as a promising candidate for the development of novel therapies that target GBM stem cells.
Collapse
Affiliation(s)
- Sharmistha Chakraborty
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Megan Tran
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Robert A Newman
- Phoenix Biotechnology, San Antonio, Texas 78217, United States
| | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.
| |
Collapse
|
333
|
Dalavaye N, Nicholas M, Pillai M, Erridge S, Sodergren MH. The Clinical Translation of α-humulene - A Scoping Review. PLANTA MEDICA 2024; 90:664-674. [PMID: 38626911 PMCID: PMC11254484 DOI: 10.1055/a-2307-8183] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 04/16/2024] [Indexed: 07/19/2024]
Abstract
α-humulene, a sesquiterpene found in essential oils of various plant species, has garnered interest due to its potential therapeutic applications. This scoping review aims to consolidate α-humulene's evidence base, informing clinical translation, and guiding future research directions. A scoping review was conducted of EMBASE, MEDLINE, and PubMed databases up to 14th July 2023. All studies describing original research on α-humulene extraction, as well as pre-clinical and clinical research, were included for review. Three hundred and forty articles were analysed. α-humulene yields ranged from negligible to 60.90% across plant species. In vitro experiments demonstrated cytotoxicity against adenocarcinomas (such as colorectal, pulmonary, breast, prostatic, lung, and ovarian), with varying responses in other cell models. Mechanistic insights revealed its involvement in mitochondrial dysfunction, diminished intracellular glutathione levels, and the induction of oxidative stress. In rodent studies, oral administration of α-humulene at 50 mg/kg reduced inflammation markers in paw oedema and ovalbumin-induced airway inflammation. Intraperitoneal administration of α-humulene (50 - 200 mg/kg) exhibited cannabimimetic properties through cannabinoid 1 and adenosine A2a receptors. α-humulene also exhibited a multitude of properties with potential scope for therapeutic utilisation. However, there is a paucity of studies that have successfully translated this research into clinical populations with the associated disease. Potential barriers to clinical translation were identified, including yield variability, limited isolation studies, and challenges associated with terpene bioavailability. Consequently, rigorous pharmacokinetic studies and further mechanistic investigations are warranted to effectively uncover the potential of α-humulene.
Collapse
Affiliation(s)
- Nishaanth Dalavaye
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Martha Nicholas
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Manaswini Pillai
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Simon Erridge
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
- Curaleaf Clinic, London, UK
| | - Mikael H. Sodergren
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
- Curaleaf International, London, UK
| |
Collapse
|
334
|
Kavipriya R, Ramasubburayan R. Phytofabrication of biocompatible zinc oxide nanoparticle using Gymnema sylvestre and its potent in vitro antibacterial, antibiofilm, and cytotoxicity against human breast cancer cells (MDA-MB-231). Bioprocess Biosyst Eng 2024; 47:1377-1391. [PMID: 38819452 DOI: 10.1007/s00449-024-03035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The increasing incidence of breast cancer and bacterial biofilm in medical devices significantly heightens global mortality and morbidity, challenging synthetic drugs. Consequently, greener-synthesized nanomaterials have emerged as a versatile alternative for various biomedical applications, offering new therapeutic avenues. This study explores the synthesis of biocompatible zinc oxide (ZnONPs) nanoparticles using Gymnema sylvestre and its antibacterial, antibiofilm, and cytotoxic properties. Characterization of ZnONPs inferred that UV-Vis spectra exhibited a sharp peak at 370 nm. Fourier transform infrared spectroscopical analysis revealed the presence of active functional groups such as aldehyde, alkyne, cyclic alkene, sulfate, alkyl aryl ether, and Zn-O bonds. X-ray diffraction analysis results confirmed the crystalline nature of the nanoparticle. Scanning electron microscope analysis evidenced hexagonal morphology, and energy-dispersive X-ray analysis confirmed zinc content. High-resolution transmission electron microscope analysis showed hexagonal and rod-shaped ZnONPs with a size of 5 nm. Zeta potential results affirmed the stability of nanoparticles. The ZnONPs effectively inhibited gram-positive (18-20 mm) than gram-negative (12-18 mm) bacterial pathogens with lower bacteriostatic and higher bactericidal values. Biofilm inhibitory property inferred ZnONPs were more effective against gram-positive (38-94%) than gram-negative bacteria (27-86%). The concentration of ZnONPs to exert 50% biofilm-inhibitory is lower against gram-positive bacteria (179.26-203.95 μg/mL) than gram-negative bacteria (201.46-236.19 μg/mL). Microscopic visualization inferred that at 250 μg/mL, ZnONPs strongly disrupted biofilm formation, as evidenced by decreased biofilm density and altered architecture. The cytotoxicity of ZnONPs against breast cancer cells showed a dose-dependent reduction in cell viability with an IC50 value of 19.4 µg/mL. AO/EB staining indicated early and late apoptotic cell death of breast cancer cells under fluorescence microscopy. The results of hemolytic activity validated the biocompatibility of the ZnONPs. Thus, the unique properties of the green-synthesized ZnONPs suggest their potential as effective drug carriers for targeted delivery in cancer therapy and the treatment of biofilm-related infections.
Collapse
Affiliation(s)
- R Kavipriya
- Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - R Ramasubburayan
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India.
| |
Collapse
|
335
|
Lee D, Im J, Kim AR, Jun W, Yun CH, Han SH. Enterococcus Phage vB_EfaS_HEf13 as an Anti-Biofilm Agent Against Enterococcus faecalis. J Microbiol 2024; 62:683-693. [PMID: 38935316 DOI: 10.1007/s12275-024-00150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Enterococcus faecalis is a Gram-positive bacterium that is frequently found in the periapical lesion of patients with apical periodontitis. Its biofilm formation in root canal is closely related to the development of refractory apical periodontitis by providing increased resistance to endodontic treatments. Phage therapy has recently been considered as an efficient therapeutic strategy in controlling various periodontal pathogens. We previously demonstrated the bactericidal capacities of Enterococcus phage vB_EfaS_HEf13 (phage HEf13) against clinically-isolated E. faecalis strains. Here, we investigated whether phage HEf13 affects biofilm formation and pre-formed biofilm of clinically-isolated E. faecalis, and its combinatory effect with endodontic treatments, including chlorhexidine (CHX) and penicillin. The phage HEf13 inhibited biofilm formation and disrupted pre-formed biofilms of E. faecalis in a dose- and time-dependent manner. Interestingly, phage HEf13 destroyed E. faecalis biofilm exopolysaccharide (EPS), which is known to be a major component of bacterial biofilm. Furthermore, combined treatment of phage HEf13 with CHX or penicillin more potently inhibited biofilm formation and disrupted pre-formed biofilm than either treatment alone. Confocal laser scanning microscopic examination demonstrated that these additive effects of the combination treatments on disruption of pre-formed biofilm are mediated by relatively enhanced reduction in thickness distribution and biomass of biofilm. Collectively, our results suggest that the effect of phage HEf13 on E. faecalis biofilm is mediated by its EPS-degrading property, and its combination with endodontic treatments more potently suppresses E. faecalis biofilm, implying that phage HEf13 has potential to be used as a combination therapy against E. faecalis infections.
Collapse
Affiliation(s)
- Dongwook Lee
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - A Reum Kim
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Woohyung Jun
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
336
|
Sivarajan K, Ravindhiran R, Sekar JN, Murugesan R, Chidambaram K, Dhandapani K. Deciphering the impact of Acinetobacter baumannii on human health, and exploration of natural compounds as efflux pump inhibitors to treat multidrug resistance. J Med Microbiol 2024; 73. [PMID: 39212030 DOI: 10.1099/jmm.0.001867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Acinetobacter baumannii is an ESKAPE pathogen and threatens human health by generating infections with high fatality rates. A. baumannii leads to a spectrum of infections such as skin and wound infections, endocarditis, meningitis pneumonia, septicaemia and urinary tract infections. Recently, strains of A. baumannii have emerged as multidrug-resistant (MDR), meaning they are resistant to at least three different classes of antibiotics. MDR development is primarily intensified by widespread antibiotic misuse and inadequate stewardship. The World Health Organization (WHO) declared A. baumannii a precarious MDR species. A. baumannii maintains the MDR phenotype via a diverse array of antimicrobial metabolite-hydrolysing enzymes, efflux of antibiotics, impermeability and antibiotic target modification, thereby complicating treatment. Hence, a deeper understanding of the resistance mechanisms employed by MDR A. baumannii can give possible approaches to treat antimicrobial resistance. Resistance-nodulation-cell division (RND) efflux pumps have been identified as the key contributors to MDR determinants, owing to their capacity to force a broad spectrum of chemical substances out of the bacterial cell. Though synthetic inhibitors have been reported previously, their efficacy and safety are of debate. As resistance-modifying agents, phytochemicals are ideal choices. These natural compounds could eliminate the bacteria or interact with pathogenicity events and reduce the bacteria's ability to evolve resistance. This review aims to highlight the mechanism behind the multidrug resistance in A. baumannii and elucidate the utility of natural compounds as efflux pump inhibitors to deal with the infections caused by A. baumannii.
Collapse
Affiliation(s)
- Karthiga Sivarajan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Ramya Ravindhiran
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Jothi Nayaki Sekar
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Rajeswari Murugesan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, School of Pharmacy, King Khalid University, Abha 652529, Saudi Arabia
| | - Kavitha Dhandapani
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| |
Collapse
|
337
|
Schuermans S, Kestens C, Marques PE. Systemic mechanisms of necrotic cell debris clearance. Cell Death Dis 2024; 15:557. [PMID: 39090111 PMCID: PMC11294570 DOI: 10.1038/s41419-024-06947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Necrosis is an overarching term that describes cell death modalities caused by (extreme) adverse conditions in which cells lose structural integrity. A guaranteed consequence of necrosis is the production of necrotic cell remnants, or debris. Necrotic cell debris is a strong trigger of inflammation, and although inflammatory responses are required for tissue healing, necrotic debris may lead to uncontrolled immune responses and collateral damage. Besides local phagocytosis by recruited leukocytes, there is accumulating evidence that extracellular mechanisms are also involved in necrotic debris clearance. In this review, we focused on systemic clearance mechanisms present in the bloodstream and vasculature that often cooperate to drive the clearance of cell debris. We reviewed the contribution and cooperation of extracellular DNases, the actin-scavenger system, the fibrinolytic system and reticuloendothelial cells in performing clearance of necrotic debris. Moreover, associations of the (mis)functioning of these clearance systems with a variety of diseases were provided, illustrating the importance of the mechanisms of clearance of dead cells in the organism.
Collapse
Affiliation(s)
- Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Caine Kestens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
338
|
Ni K, Meng L. Mechanism of PANoptosis in metabolic dysfunction-associated steatotic liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102381. [PMID: 38821484 DOI: 10.1016/j.clinre.2024.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
In recent years, the incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) has been steadily rising, emerging as a major chronic liver disease of global concern. The course of MASLD is varied, spanning from MASLD to metabolic dysfunction associated steatohepatitis (MASH). MASH is an important contributor to cirrhosis, which may subsequently lead to hepatocellular carcinoma. It has been found that PANoptosis, an emerging inflammatory programmed cell death (PCD), is involved in the pathogenesis of MASLD and facilitates the development of NASH, eventually resulting in inflammatory fibrosis and hepatocyte death. This paper reviews the latest research progress on PANoptosis and MASLD to understand the mechanism of MASLD and provide new directions for future treatment and drug development.
Collapse
Affiliation(s)
- Keying Ni
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medical), Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou, China
| | - Lina Meng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medical), Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
339
|
Malaikozhundan B, Mohandoss S, Krishnamoorthi R, Bharathi PV, Palanisamy S, Vinodhini J. Enhanced bactericidal, antibiofilm and antioxidative response of Lawsonia inermis leaf extract synthesized ZnO NPs loaded with commercial antibiotic. Bioprocess Biosyst Eng 2024; 47:1241-1257. [PMID: 38607416 DOI: 10.1007/s00449-024-03000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
Globally, antibiotic resistance is a challenging issue in healthcare sector. The emergence of multiple drug-resistant bacteria has forced us to modify existing medicines and or formulate newer medicines that are effective and inexpensive. In this perspective, this study involves the formation of zinc oxide nanoparticles (ZnO NPs) by utilizing the Lawsonia inermis (Li) leaf extract. The prepared L. inermis leaf extract mediated ZnO NPs (Li-ZnO NPs) were bio-physically characterized. The antibacterial and radical scavenging effects of Li-ZnO NPs were evaluated. In addition, ZnO NPs were conjugated with standard antibiotic (ciprofloxacin) and its drug loading efficiency, drug release and antibacterial efficacy were tested and compared with non-drug loaded ZnO NPs. An absorbance peak at 340 nm was noted for Li-ZnO NPs. After conjugation with the drug, two absorbance peaks- one at 242 nm characteristic of ciprofloxacin and the other at 350 nm characteristics of ZnO NPs were observed. The crystallite size was 18.7 nm as determined by XRD. The antibacterial effect was higher on Gram-positive (S. aureus and S. pyogenes) than the Gram-negative pathogens (E. coli and K. pneumoniae). Inhibition of S. aureus and S. pyogenes biofilm at 100 μg mL-1were, respectively, 97.5 and 92.6%. H2O2 free radicals was inhibited to 90% compared to the standard ascorbic acid at 100 μg mL-1. After drug loading, the FTIR spectrum confirmed the existence of ciprofloxacin peaks at 965 cm-1 and Zn-O bond at 492 cm-1. The drug loading capacity of 15 nm sized ZnO NPs was higher (58, 75, 90 and 95% at 1, 2.5, 5 and 10% drug concentrations, respectively) compared to 20 nm. Similarly, the percentage of drug (ciprofloxacin) released from 15 nm ZnO NPs were increased to 90% at 10% drug-loaded samples, respectively. Also, the antibiotic loaded ZnO NPs had significant antibacterial effects against tested bacteria compared to Li-ZnO NPs and ciprofloxacin alone. This revealed that the antibiotic loaded ZnO NPs offer a sustainable route to treat multi-drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Balasubramanian Malaikozhundan
- Department of Biology, The Gandhigram Rural Institute (Deemed to Be University), 624 302, Gandhigram, Dindigul District, Tamil Nadu, India.
| | - Sonaimuthu Mohandoss
- School of Chemical Engineering, Yeungnam University, 38541, Gyeongsan, Republic of Korea
| | - Raman Krishnamoorthi
- Department of Biology, The Gandhigram Rural Institute (Deemed to Be University), 624 302, Gandhigram, Dindigul District, Tamil Nadu, India
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 333, Taoyuan, Taiwan
| | - Palanichamy Vidhya Bharathi
- Department of Biology, The Gandhigram Rural Institute (Deemed to Be University), 624 302, Gandhigram, Dindigul District, Tamil Nadu, India
| | - Subramanian Palanisamy
- East Coast Life Sciences Institute, Gangneung-Wonju National University, 120, Gangneung, 210-702, Gangwon, Republic of Korea
| | - Jayaraj Vinodhini
- Department of Biotechnology, Dr. Umayal Ramanathan College for Women, Alagappa University, 630 003, Karaikudi, Tamil Nadu, India
- Parvathy's Anugrahaa International School, 624 002, Dindigul, Tamil Nadu, India
| |
Collapse
|
340
|
Galis P, Bartosova L, Farkasova V, Bartekova M, Ferenczyova K, Rajtik T. Update on clinical and experimental management of diabetic cardiomyopathy: addressing current and future therapy. Front Endocrinol (Lausanne) 2024; 15:1451100. [PMID: 39140033 PMCID: PMC11319149 DOI: 10.3389/fendo.2024.1451100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe secondary complication of type 2 diabetes mellitus (T2DM) that is diagnosed as a heart disease occurring in the absence of any previous cardiovascular pathology in diabetic patients. Although it is still lacking an exact definition as it combines aspects of both pathologies - T2DM and heart failure, more evidence comes forward that declares DCM as one complex disease that should be treated separately. It is the ambiguous pathological phenotype, symptoms or biomarkers that makes DCM hard to diagnose and screen for its early onset. This re-view provides an updated look on the novel advances in DCM diagnosis and treatment in the experimental and clinical settings. Management of patients with DCM proposes a challenge by itself and we aim to help navigate and advice clinicians with early screening and pharmacotherapy of DCM.
Collapse
Affiliation(s)
- Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Kristina Ferenczyova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
341
|
Rabiei M, McAllister MM, Gassman NR, Lee KJ, Acton S, Liebhart D, Low WY, Hemmatzadeh F. Newcastle Disease Virus Induces Profound Lymphoid Depletion with Different Patterns of Necroptosis, Necrosis, and Oxidative DNA Damage in Bursa, Spleen, and Other Lymphoid Tissues. Pathogens 2024; 13:619. [PMID: 39204220 PMCID: PMC11357213 DOI: 10.3390/pathogens13080619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
This study delves into the pathogenesis of virulent genotype VII strains of the Newcastle disease virus (NDV), focusing on experimentally infected birds. Predominant and consistent lesions observed include bursal atrophy and extensive depletion of all lymphoid tissues. Immunohistochemistry (IHC) analysis, targeting apoptosis (Caspase-3), necroptosis (MLKL), and NDV markers, indicates that bursal atrophy is linked to a non-apoptotic programmed cell death pathway known as "necroptosis". Repair assisted damage detection (RADD) of the bursa reveal oxidative DNA damage patterns consistent with programmed cell death, aligning with MLKL expression. Contrastingly, in the spleen, our findings suggest that necrosis (non-programmed cell death) predominantly contributes to lymphoid depletion. This conclusion is supported by evidence of karyorrhexis, fibrinous inflammation, RADD analyses, and IHC. Moreover, in addition to being pathogenic in its own right, NDV caused extensive and rapid lymphoid depletion that should be expected to contribute to profound immunosuppression. The elucidation of necroptosis in NDV-infected chickens provides a good rationale to investigate this mechanism in other paramyxoviral diseases such as human measles.
Collapse
Affiliation(s)
- Mohammad Rabiei
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
- Gastro-Intestinal Viral Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, Sydney, NSW 2170, Australia
| | - Milton M. McAllister
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
| | - Natalie R. Gassman
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Kevin J. Lee
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Sydney Acton
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Dieter Liebhart
- Clinical Department for Farm Animals and Food System Science, Clinical Centre for Population Medicine in Fish, Pig and Poultry, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Wai Yee Low
- The Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
| | - Farhid Hemmatzadeh
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
- The Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
| |
Collapse
|
342
|
Mohamed H, Marusich E, Divashuk M, Leonov S. A unique combination of natural fatty acids from Hermetia illucens fly larvae fat effectively combats virulence factors and biofilms of MDR hypervirulent mucoviscus Klebsiella pneumoniae strains by increasing Lewis acid-base/van der Waals interactions in bacterial wall membranes. Front Cell Infect Microbiol 2024; 14:1408179. [PMID: 39119288 PMCID: PMC11306206 DOI: 10.3389/fcimb.2024.1408179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Hypervirulent Klebsiella pneumoniae (hvKp) and carbapenem-resistant K. pneumoniae (CR-Kp) are rapidly emerging as opportunistic pathogens that have a global impact leading to a significant increase in mortality rates among clinical patients. Anti-virulence strategies that target bacterial behavior, such as adhesion and biofilm formation, have been proposed as alternatives to biocidal antibiotic treatments to reduce the rapid emergence of bacterial resistance. The main objective of this study was to examine the efficacy of fatty acid-enriched extract (AWME3) derived from the fat of Black Soldier Fly larvae (Hermetia illucens) in fighting against biofilms of multi-drug resistant (MDR) and highly virulent Klebsiella pneumoniae (hvKp) pathogens. Additionally, the study also aimed to investigate the potential mechanisms underlying this effect. Methods Crystal violet (CV) and ethidium bromide (EtBr) assays show how AWME3 affects the formation of mixed and mature biofilms by the KP ATCC BAA-2473, KPi1627, and KPM9 strains. AWME3 has shown exceptional efficacy in combating the hypermucoviscosity (HMV) virulent factors of KPi1627 and KPM9 strains when tested using the string assay. The rudimentary motility of MDR KPM9 and KP ATCC BAA-2473 strains was detected through swimming, swarming, and twitching assays. The cell wall membrane disturbances induced by AWME3 were detected by light and scanning electron microscopy and further validated by an increase in the bacterial cell wall permeability and Lewis acid-base/van der Waals characteristics of K. pneumoniae strains tested by MATS (microbial adhesion to solvents) method. Results After being exposed to 0.5 MIC (0.125 mg/ml) of AWME3, a significant reduction in the rudimentary motility of MDR KPM9 and KP ATCC BAA-2473 strains, whereas the treated bacterial strains exhibited motility between 4.23 ± 0.25 and 4.47 ± 0.25 mm, while the non-treated control groups showed significantly higher motility ranging from 8.5 ± 0.5 to 10.5 ± 0.5 mm. Conclusion In conclusion, this study demonstrates the exceptional capability of the natural AWME3 extract enriched with a unique combination of fatty acids to effectively eliminate the biofilms formed by the highly drug-resistant and highly virulent K. pneumoniae (hvKp) pathogens. Our results highlight the opportunity to control and minimize the rapid emergence of bacterial resistance through the treatment using AWME3 of biofilm-associated infections caused by hvKp and CRKp pathogens.
Collapse
Affiliation(s)
- Heakal Mohamed
- Agricultural Research Center (ARC), Plant Protection Research Institute (PPRI), Giza, Egypt
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
| | - Elena Marusich
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
| | - Mikhail Divashuk
- All-Russia Research Institute of Agricultural Biotechnology Kurchatov Genomic Center - VNIISB, Moscow, Russia
| | - Sergey Leonov
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
343
|
Park J, Xiang Z, Liu Y, Li CH, Chen C, Nagaraj H, Nguyen T, Nabawy A, Koo H, Rotello VM. Surface-Charge Tuned Polymeric Nanoemulsions for Carvacrol Delivery in Interkingdom Biofilms. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37613-37622. [PMID: 39007413 PMCID: PMC11624604 DOI: 10.1021/acsami.4c06618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Biofilms, intricate microbial communities entrenched in extracellular polymeric substance (EPS) matrices, pose formidable challenges in infectious disease treatment, especially in the context of interkingdom biofilms prevalent in the oral environment. This study investigates the potential of carvacrol-loaded biodegradable nanoemulsions (NEs) with systematically varied surface charges─cationic guanidinium (GMT-NE) and anionic carboxylate (CMT-NE). Zeta potentials of +25 mV (GMT-NE) and -33 mV (CMT-NE) underscore successful nanoemulsion fabrication (∼250 nm). Fluorescent labeling and dynamic tracking across three dimensions expose GMT-NE's superior diffusion into oral biofilms, yielding a robust antimicrobial effect with 99.99% killing for both streptococcal and Candida species and marked reductions in bacterial cell viability compared to CMT-NE (∼4-log reduction). Oral mucosa tissue cultures affirm the biocompatibility of both NEs with no morphological or structural changes, showcasing their potential for combating intractable biofilm infections in oral environment. This study advances our understanding of NE surface charges and their interactions within interkingdom biofilms, providing insights crucial for addressing complex infections involving bacteria and fungi in the demanding oral context.
Collapse
Affiliation(s)
- Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Zhenting Xiang
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, 240 S 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Yuan Liu
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Preventive & Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cheng-Hsuan Li
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Tiffany Nguyen
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Hyun Koo
- Biofilm Research Laboratories, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, 240 S 40th Street, Philadelphia, Pennsylvania 19104, United States
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, 240 S 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
344
|
Nguyen NTT, Müller R, Briukhovetska D, Weber J, Feucht J, Künkele A, Hudecek M, Kobold S. The Spectrum of CAR Cellular Effectors: Modes of Action in Anti-Tumor Immunity. Cancers (Basel) 2024; 16:2608. [PMID: 39061247 PMCID: PMC11274444 DOI: 10.3390/cancers16142608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor-T cells have spearheaded the field of adoptive cell therapy and have shown remarkable results in treating hematological neoplasia. Because of the different biology of solid tumors compared to hematological tumors, response rates of CAR-T cells could not be transferred to solid entities yet. CAR engineering has added co-stimulatory domains, transgenic cytokines and switch receptors to improve performance and persistence in a hostile tumor microenvironment, but because of the inherent cell type limitations of CAR-T cells, including HLA incompatibility, toxicities (cytokine release syndrome, neurotoxicity) and high costs due to the logistically challenging preparation process for autologous cells, the use of alternative immune cells is gaining traction. NK cells and γδ T cells that do not need HLA compatibility or macrophages and dendritic cells with additional properties such as phagocytosis or antigen presentation are increasingly seen as cellular vehicles with potential for application. As these cells possess distinct properties, clinicians and researchers need a thorough understanding of their peculiarities and commonalities. This review will compare these different cell types and their specific modes of action seen upon CAR activation.
Collapse
Affiliation(s)
- Ngoc Thien Thu Nguyen
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (N.T.T.N.); (R.M.); (D.B.)
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between the DKFZ Heidelberg and the University Hospital of the LMU, 80336 Munich, Germany
| | - Rasmus Müller
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (N.T.T.N.); (R.M.); (D.B.)
| | - Daria Briukhovetska
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (N.T.T.N.); (R.M.); (D.B.)
| | - Justus Weber
- Department of Medicine II, Chair in Cellular Immunotherapy, University Hospital Würzburg, 97080 Würzburg, Germany; (J.W.); (M.H.)
| | - Judith Feucht
- Cluster of Excellence iFIT “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tuebingen, Germany;
- Department of Hematology and Oncology, University Children’s Hospital Tuebingen, University of Tübingen, 72076 Tuebingen, Germany
| | - Annette Künkele
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany;
- German Cancer Consortium (DKTK), Partner Site Berlin, 10117 Berlin, Germany
| | - Michael Hudecek
- Department of Medicine II, Chair in Cellular Immunotherapy, University Hospital Würzburg, 97080 Würzburg, Germany; (J.W.); (M.H.)
- Fraunhofer Institute for Cell Therapy and Immunology, Cellular Immunotherapy Branch Site Würzburg, 97080 Würzburg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (N.T.T.N.); (R.M.); (D.B.)
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between the DKFZ Heidelberg and the University Hospital of the LMU, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München—German Research Center for Environmental Health Neuherberg, 85764 Oberschleißheim, Germany
| |
Collapse
|
345
|
Sahu K, Kurrey R, Pillai AK. Green synthesis of silver nanoparticles from Manilkara zapota leaf extract for the detection of aminoglycoside antibiotics and other applications. RSC Adv 2024; 14:23240-23256. [PMID: 39045403 PMCID: PMC11265568 DOI: 10.1039/d4ra01906g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024] Open
Abstract
Antibiotics of aminoglycoside (AMG) class, such as streptomycin (STR), have been widely used to treat infectious diseases caused by Gram-negative bacteria in livestock and humans. In this study, a selective and sensitive colorimetric probe for the determination of STR was proposed based on eco-friendly green synthesized AgNPs from the leaf extract of Manilkara zapota. The mechanism for the detection of STR is based on the electrostatic interaction of opposite charges between negatively charged silver nanoparticle-Manilkara zapota leaf (AgNP-MZL) and STR, causing an aggregation-induced characteristic shift of the SPR band (from 390 nm to 570 nm wavelength) of AgNP-MZL. The morphology, size distribution and optical properties of AgNP-MZL were characterized using UV/visible absorption spectroscopy, FTIR spectroscopy, XRD, DLS, zeta-potential measurements and TEM. The selective determination of STR was experimentally confirmed by performing controlled testing with other classes of antibiotics. To test the sensitivity level of this method, the ratio of these two A 390/A 570 absorbance wavelengths was selected to provide a linear concentration plot between 5 and 100 ng mL-1 STR. The LOD and LOQ were calculated to be 3.5 ng mL-1 and 26.8 ng mL-1, respectively. Good precision was evaluated with a standard deviation of 0.45 ng mL-1 and a relative standard deviation of 2.0% (intraday) and 2.42% (interday) at 10 ng mL-1 for 3 replicate measurements. Advantages of the green synthesis of AgNP-MZL include its eco-friendly nature and it is easy, efficient, cost effective and selective for the detection of the AMG class of antibiotics, i.e. STR, in agricultural and environmental samples.
Collapse
Affiliation(s)
- Khushboo Sahu
- Govt. V. Y. T. Post Graduate Autonomous College Durg-491 001 Chhattisgarh India +917882 393644
| | - Ramsingh Kurrey
- National Center for Natural Resources, Pt. Ravishankar Shukla University Raipur-492 010 Chhattisgarh India
| | - Ajai Kumar Pillai
- Govt. V. Y. T. Post Graduate Autonomous College Durg-491 001 Chhattisgarh India +917882 393644
| |
Collapse
|
346
|
Yang S, Meng X, Zhen Y, Baima Q, Wang Y, Jiang X, Xu Z. Strategies and mechanisms targeting Enterococcus faecalis biofilms associated with endodontic infections: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1433313. [PMID: 39091674 PMCID: PMC11291369 DOI: 10.3389/fcimb.2024.1433313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.
Collapse
Affiliation(s)
- Shipeng Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuqi Zhen
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Quzhen Baima
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinmiao Jiang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhibo Xu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
347
|
Ullah N, Hasnain SZU, Baloch R, Amin A, Nasibova A, Selakovic D, Rosic GL, Islamov S, Naraliyeva N, Jaradat N, Mammadova AO. Exploring essential oil-based bio-composites: molecular docking and in vitro analysis for oral bacterial biofilm inhibition. Front Chem 2024; 12:1383620. [PMID: 39086984 PMCID: PMC11288909 DOI: 10.3389/fchem.2024.1383620] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/15/2024] [Indexed: 08/02/2024] Open
Abstract
Oral bacterial biofilms are the main reason for the progression of resistance to antimicrobial agents that may lead to severe conditions, including periodontitis and gingivitis. Essential oil-based nanocomposites can be a promising treatment option. We investigated cardamom, cinnamon, and clove essential oils for their potential in the treatment of oral bacterial infections using in vitro and computational tools. A detailed analysis of the drug-likeness and physicochemical properties of all constituents was performed. Molecular docking studies revealed that the binding free energy of a Carbopol 940 and eugenol complex was -2.0 kcal/mol, of a Carbopol 940-anisaldehyde complex was -1.9 kcal/mol, and a Carbapol 940-eugenol-anisaldehyde complex was -3.4 kcal/mol. Molecular docking was performed against transcriptional regulator genes 2XCT, 1JIJ, 2Q0P, 4M81, and 3QPI. Eugenol cinnamaldehyde and cineol presented strong interaction with targets. The essential oils were analyzed against Staphylococcus aureus and Staphylococcus epidermidis isolated from the oral cavity of diabetic patients. The cinnamon and clove essential oil combination presented significant minimum inhibitory concentrations (MICs) (0.0625/0.0312 mg/mL) against S. epidermidis and S. aureus (0.0156/0.0078 mg/mL). In the anti-quorum sensing activity, the cinnamon and clove oil combination presented moderate inhibition (8 mm) against Chromobacterium voilaceum with substantial violacein inhibition (58% ± 1.2%). Likewise, a significant biofilm inhibition was recorded in the case of S. aureus (82.1% ± 0.21%) and S. epidermidis (84.2% ± 1.3%) in combination. It was concluded that a clove and cinnamon essential oil-based formulation could be employed to prepare a stable nanocomposite, and Carbapol 940 could be used as a compatible biopolymer.
Collapse
Affiliation(s)
- Niamat Ullah
- Natural Products Research Lab, Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Syed Zia Ul Hasnain
- Department of Pharmacognosy, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Rabia Baloch
- Allama Iqbal Teaching Hospital, Dera Ghazi Khan, Pakistan
| | - Adnan Amin
- Natural Products Research Lab, Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Aygun Nasibova
- Department of Biophysics and Biochemistry, Baku State University, Baku, Azerbaijan
- Institute of Radiation Problems, Ministry of Science and Education Republic of Azerbaijan, Baku, Azerbaijan
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gvozden Luka Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Sokhib Islamov
- Department of Technology of Storage and Processing of Agricultural Products, Tashkent State Agrarian University, Tashkent, Uzbekistan
| | | | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | |
Collapse
|
348
|
Tang D, Lin Y, Yao H, Liu Y, Xi Y, Li M, Mao A. Effect of L-HSL on biofilm and motility of Pseudomonas aeruginosa and its mechanism. Appl Microbiol Biotechnol 2024; 108:418. [PMID: 39012538 PMCID: PMC11252199 DOI: 10.1007/s00253-024-13247-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) biofilm formation is a crucial cause of enhanced antibiotic resistance. Quorum sensing (QS) is involved in regulating biofilm formation; QS inhibitors block the QS signaling pathway as a new strategy to address bacterial resistance. This study investigated the potential and mechanism of L-HSL (N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide) as a QS inhibitor for P. aeruginosa. The results showed that L-HSL effectively inhibited the biofilm formation and dispersed the pre-formed biofilm of P. aeruginosa. The production of extracellular polysaccharides and the motility ability of P. aeruginosa were suppressed by L-HSL. C. elegans infection experiment showed that L-HSL was non-toxic and provided protection to C. elegans against P. aeruginosa infection. Transcriptomic analysis revealed that L-HSL downregulated genes related to QS pathways and biofilm formation. L-HSL exhibits a promising potential as a therapeutic drug for P. aeruginosa infection. KEY POINTS: • Chemical synthesis of N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide, named L-HSL. • L-HSL does not generate survival pressure on the growth of P. aeruginosa and can inhibit the QS system. • KEGG enrichment analysis found that after L-HSL treatment, QS-related genes were downregulated.
Collapse
Affiliation(s)
- Deping Tang
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yanyan Lin
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Huihui Yao
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yali Liu
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yanpeng Xi
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Mengjiao Li
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Aihong Mao
- Gansu Provincial Academic Institute for Medical Research, Lanzhou, 730050, Gansu, China.
| |
Collapse
|
349
|
Boreak N, Al Mahde RZ, Otayn WA, Alamer AY, Alrajhi T, Jafri S, Sharwani A, Swaidi E, Abozoah S, Mowkly AAM. Exploring Plant-Based Compounds as Alternatives for Targeting Enterococcus faecalis in Endodontic Therapy: A Molecular Docking Approach. Int J Mol Sci 2024; 25:7727. [PMID: 39062969 PMCID: PMC11276846 DOI: 10.3390/ijms25147727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Endodontic infections pose significant challenges in dental practice due to their persistence and potential complications. Among the causative agents, Enterococcus faecalis stands out for its ability to form biofilms and develop resistance to conventional antibiotics, leading to treatment failures and recurrent infections. The urgent need for alternative treatments arises from the growing concern over antibiotic resistance and the limitations of current therapeutic options in combating E. faecalis-associated endodontic infections. Plant-based natural compounds offer a promising avenue for exploration, given their diverse bioactive properties and potential as sources of novel antimicrobial agents. In this study, molecular docking and dynamics simulations are employed to explore the interactions between SrtA, a key enzyme in E. faecalis, and plant-based natural compounds. Analysis of phytocompounds through molecular docking unveiled several candidates with binding energies surpassing that of the control drug, ampicillin, with pinocembrin emerging as the lead compound due to its strong interactions with key residues of SrtA. Comparative analysis with ampicillin underscored varying degrees of structural similarity among the study compounds. Molecular dynamics simulations provided deeper insights into the dynamic behavior and stability of protein-ligand complexes, with pinocembrin demonstrating minimal conformational changes and effective stabilization of the N-terminal region. Free energy landscape analysis supported pinocembrin's stabilizing effects, further corroborated by hydrogen bond analysis. Additionally, physicochemical properties analysis highlighted the drug-likeness of pinocembrin and glabridin. Overall, this study elucidates the potential anti-bacterial properties of selected phytocompounds against E. faecalis infections, with pinocembrin emerging as a promising lead compound for further drug development efforts, offering new avenues for combating bacterial infections and advancing therapeutic interventions in endodontic practice.
Collapse
Affiliation(s)
- Nezar Boreak
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Rahf Zuhair Al Mahde
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Waseem Ahmed Otayn
- Specialized Dental Canter, Ministry of Health, Jazan 45142, Saudi Arabia; (W.A.O.)
| | - Amwaj Yahya Alamer
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Taif Alrajhi
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Shatha Jafri
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Amnah Sharwani
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Entesar Swaidi
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | - Shahad Abozoah
- Department of Restorative Dental Sciences, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia; (R.Z.A.M.); (A.Y.A.); (T.A.); (S.J.); (A.S.)
| | | |
Collapse
|
350
|
Lyu P, Wen J, Zhang W, Liu N, Stolzer I, Gießl A, Jia Y, Mauro D, Zhang F, Ciccia F, Soulat D, Günther C, Schett G, Bozec A. Expression of HIF1α in intestinal epithelium restricts arthritis inflammation by inhibiting RIPK3-induced cell death machinery. Ann Rheum Dis 2024; 83:984-997. [PMID: 38503474 PMCID: PMC11287550 DOI: 10.1136/ard-2023-224491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 02/28/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVES To investigate the mechanism by which intestinal epithelial cell (IEC) death induces arthritis. METHODS IEC death was assessed by staining for necroptosis and apoptosis markers and fluorescence in situ hybridisation at different time points during collagen-induced arthritis (CIA). During the development of CIA, messenger RNA (mRNA) sequencing was performed, followed by Gene Ontology enrichment analysis of differentially expressed genes. Mice deficient for hypoxia-inducible factor 1α (Hif1a) in IECs (Hif1a ∆IEC) were generated and induced for arthritis. mRNA sequencing, chromatin immunoprecipitated (ChIP) DNA sequencing and ChIP-qualitative PCR were performed on IECs from Hif1a ∆IEC mice and littermate controls. Effects of HIF1α stabilisation by inhibition of prolyl hydroxylase domain-containing enzymes and treatment with the inhibitor of receptor-interacting protein kinase-3 (RIPK3) were tested in intestinal organoids and in CIA. RESULTS IEC underwent apoptotic and necroptotic cell death at the onset of arthritis, leading to impaired gut barrier function. HIF1α was identified as one of the most upregulated genes in IECs during the onset of arthritis. Deletion of Hif1a in IEC enhanced IEC necroptosis, triggered intestinal inflammation and exacerbated arthritis. HIF1α was found to be a key transcriptional repressor for the necroptosis-inducing factor RIPK3. Enhanced RIPK3 expression, indicating necroptosis, was also found in the intestinal epithelium of patients with new-onset rheumatoid arthritis. Therapeutic stabilisation of HIF1α as well as small-molecule-based RIPK3 inhibition rescued intestinal necroptosis in vitro and in vivo and suppressed the development of arthritis. CONCLUSION Our results identify IEC necroptosis as a critical link between the gut and the development of arthritis.
Collapse
Affiliation(s)
- Pang Lyu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jinming Wen
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wenshuo Zhang
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ning Liu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iris Stolzer
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Andreas Gießl
- Department of Opthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yewei Jia
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Daniele Mauro
- Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Fulin Zhang
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Francesco Ciccia
- Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Didier Soulat
- Microbiology Institute, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Claudia Günther
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|