1
|
Nicholas-Haizelden K, Murphy B, Hoptroff M, Horsburgh MJ. Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products. Microorganisms 2023; 11:1899. [PMID: 37630459 PMCID: PMC10456854 DOI: 10.3390/microorganisms11081899] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Bioprospecting is the discovery and exploration of biological diversity found within organisms, genetic elements or produced compounds with prospective commercial or therapeutic applications. The human skin is an ecological niche which harbours a rich and compositional diversity microbiome stemming from the multifactorial interactions between the host and microbiota facilitated by exploitable effector compounds. Advances in the understanding of microbial colonisation mechanisms alongside species and strain interactions have revealed a novel chemical and biological understanding which displays applicative potential. Studies elucidating the organismal interfaces and concomitant understanding of the central processes of skin biology have begun to unravel a potential wealth of molecules which can exploited for their proposed functions. A variety of skin-microbiome-derived compounds display prospective therapeutic applications, ranging from antioncogenic agents relevant in skin cancer therapy to treatment strategies for antimicrobial-resistant bacterial and fungal infections. Considerable opportunities have emerged for the translation to personal care products, such as topical agents to mitigate various skin conditions such as acne and eczema. Adjacent compound developments have focused on cosmetic applications such as reducing skin ageing and its associated changes to skin properties and the microbiome. The skin microbiome contains a wealth of prospective compounds with therapeutic and commercial applications; however, considerable work is required for the translation of in vitro findings to relevant in vivo models to ensure translatability.
Collapse
Affiliation(s)
- Keir Nicholas-Haizelden
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| | - Barry Murphy
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Michael Hoptroff
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Malcolm J. Horsburgh
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| |
Collapse
|
2
|
Abdelhamid AG, Yousef AE. Combating Bacterial Biofilms: Current and Emerging Antibiofilm Strategies for Treating Persistent Infections. Antibiotics (Basel) 2023; 12:1005. [PMID: 37370324 DOI: 10.3390/antibiotics12061005] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Biofilms are intricate multicellular structures created by microorganisms on living (biotic) or nonliving (abiotic) surfaces. Medically, biofilms often lead to persistent infections, increased antibiotic resistance, and recurrence of infections. In this review, we highlighted the clinical problem associated with biofilm infections and focused on current and emerging antibiofilm strategies. These strategies are often directed at disrupting quorum sensing, which is crucial for biofilm formation, preventing bacterial adhesion to surfaces, impeding bacterial aggregation in viscous mucus layers, degrading the extracellular polymeric matrix, and developing nanoparticle-based antimicrobial drug complexes which target persistent cells within the biofilm core. It is important to acknowledge, however, that the use of antibiofilm agents faces obstacles, such as limited effectiveness in vivo, potential cytotoxicity to host cells, and propensity to elicit resistance in targeted biofilm-forming microbes. Emerging next generation antibiofilm strategies, which rely on multipronged approaches, were highlighted, and these benefit from current advances in nanotechnology, synthetic biology, and antimicrobial drug discovery. The assessment of current antibiofilm mitigation approaches, as presented here, could guide future initiatives toward innovative antibiofilm therapeutic strategies. Enhancing the efficacy and specificity of some emerging antibiofilm strategies via careful investigations, under conditions that closely mimic biofilm characteristics within the human body, could bridge the gap between laboratory research and practical application.
Collapse
Affiliation(s)
- Ahmed G Abdelhamid
- Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Court, Columbus, OH 43210, USA
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Ahmed E Yousef
- Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Court, Columbus, OH 43210, USA
- Department of Microbiology, The Ohio State University, 105 Biological Sciences Building, 484 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Rizzetto G, Molinelli E, Radi G, Cirioni O, Brescini L, Giacometti A, Offidani A, Simonetti O. MRSA and Skin Infections in Psoriatic Patients: Therapeutic Options and New Perspectives. Antibiotics (Basel) 2022; 11:1504. [PMID: 36358159 PMCID: PMC9686594 DOI: 10.3390/antibiotics11111504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Psoriatic patients present various infectious risk factors, but there are few studies in the literature evaluating the actual impact of psoriasis in severe staphylococcal skin infections. Our narrative review of the literature suggests that psoriatic patients are at increased risk of both colonization and severe infection, during hospitalization, by S. aureus. The latter also appears to play a role in the pathogenesis of psoriasis through the production of exotoxins. Hospitalized psoriatic patients are also at increased risk of MRSA skin infections. For this reason, new molecules are needed that could both overcome bacterial resistance and inhibit exotoxin production. In our opinion, in the near future, topical quorum sensing inhibitors in combination with current anti-MRSA therapies will be able to overcome the increasing resistance and block exotoxin production. Supplementation with Vitamin E (VE) or derivatives could also enhance the effect of anti-MRSA antibiotics, considering that psoriatic patients with metabolic comorbidities show a low intake of VE and low serum levels, making VE supplementation an interesting new perspective.
Collapse
Affiliation(s)
- Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Giulia Radi
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lucia Brescini
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| |
Collapse
|
4
|
Suda T, Hanawa T, Tanaka M, Tanji Y, Miyanaga K, Hasegawa-Ishii S, Shirato K, Kizaki T, Matsuda T. Modification of the immune response by bacteriophages alters methicillin-resistant Staphylococcus aureus infection. Sci Rep 2022; 12:15656. [PMID: 36123529 PMCID: PMC9483902 DOI: 10.1038/s41598-022-19922-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
There is an urgent need to develop phage therapies for multidrug-resistant bacterial infections. However, although bacteria have been shown to be susceptible to phage therapy, phage therapy is not sufficient in some cases. PhiMR003 is a methicillin-resistant Staphylococcus aureus phage previously isolated from sewage influent, and it has demonstrated high lytic activity and a broad host range to MRSA clinical isolates in vitro. To investigate the potential of phiMR003 for the treatment of MRSA infection, the effects of phiMR003 on immune responses in vivo were analysed using phiMR003-susceptible MRSA strains in a mouse wound infection model. Additionally, we assessed whether phiMR003 could affect the immune response to infection with a nonsusceptible MRSA strain. Interestingly, wounds infected with both susceptible and nonsusceptible MRSA strains treated with phiMR003 demonstrated decreased bacterial load, reduced inflammation and accelerated wound closure. Moreover, the infiltration of inflammatory cells in infected tissue was altered by phiMR003. While the effects of phiMR003 on inflammation and bacterial load disappeared with heat inactivation of phiMR003. Transcripts of proinflammatory cytokines induced by lipopolysaccharide were reduced in mouse peritoneal macrophages. These results show that the immune modulation occurring as a response to the phage itself improves the clinical outcomes of phage therapy.
Collapse
Affiliation(s)
- Tomoya Suda
- Department of General Medicine, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Tomoko Hanawa
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo, 181-8611, Japan.
| | - Mayuko Tanaka
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Yasunori Tanji
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 J3-8 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 J3-8 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan.,Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Sanae Hasegawa-Ishii
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| | - Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Takeaki Matsuda
- Department of General Medicine, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo, 181-8611, Japan. .,Department of Traumatology and Critical Care Medicine, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo, 181-8611, Japan.
| |
Collapse
|
5
|
Role of Daptomycin in Cutaneous Wound Healing: A Narrative Review. Antibiotics (Basel) 2022; 11:antibiotics11070944. [PMID: 35884198 PMCID: PMC9311791 DOI: 10.3390/antibiotics11070944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
Daptomycin is active against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA) and the on-label indications for its use include complicated skin and skin structure infections (cSSSI). We performed a narrative review of the literature with the aim to evaluate the role of daptomycin in the skin wound healing process, proposing our point of view on the possible association with other molecules that could improve the skin healing process. Daptomycin may improve wound healing in MRSA-infected burns, surgical wounds, and diabetic feet, but further studies in humans with histological examination are needed. In the future, the combination of daptomycin with other molecules with synergistic action, such as vitamin E and derivates, IB-367, RNA III-inhibiting peptide (RIP), and palladium nanoflowers, may help to improve wound healing and overcome forms of antibiotic resistance.
Collapse
|
6
|
Recent Strategies to Combat Biofilms Using Antimicrobial Agents and Therapeutic Approaches. Pathogens 2022; 11:pathogens11030292. [PMID: 35335616 PMCID: PMC8955104 DOI: 10.3390/pathogens11030292] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Biofilms are intricate bacterial assemblages that attach to diverse surfaces using an extracellular polymeric substance that protects them from the host immune system and conventional antibiotics. Biofilms cause chronic infections that result in millions of deaths around the world every year. Since the antibiotic tolerance mechanism in biofilm is different than that of the planktonic cells due to its multicellular structure, the currently available antibiotics are inadequate to treat biofilm-associated infections which have led to an immense need to find newer treatment options. Over the years, various novel antibiofilm compounds able to fight biofilms have been discovered. In this review, we have focused on the recent and intensively researched therapeutic techniques and antibiofilm agents used for biofilm treatment and grouped them according to their type and mode of action. We also discuss some therapeutic approaches that have the potential for future advancement.
Collapse
|
7
|
Asaoka K, Sasaki K, Yagi J, Takahashi F, Nagano K, Yamamoto T, Sugano I, Okano T, Hasumi M, Nakatani Y, Takano T, Yamasaki Y, Ookawa J, Kunishima H. Alleviation of hand-skin roughness after use of alcohol-based hand rub with inhibitory effects on Staphylococcus aureus–producing δ-toxin. J Infect Chemother 2022; 28:684-689. [DOI: 10.1016/j.jiac.2022.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/05/2022] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
|
8
|
Simonetti O, Rizzetto G, Cirioni O, Molinelli E, Morroni G, Giacometti A, Offidani A. New insight into old and new antimicrobial molecules targeting quorum sensing for MRSA wound infection. Future Microbiol 2022; 17:177-183. [PMID: 35040689 DOI: 10.2217/fmb-2021-0120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
MRSA represents one of the largest problems in wound healing as a result of its increasing incidence and the complex therapeutic approach required to treat it. The need for new solutions to overcome antibiotic resistance led to the development of antimicrobial molecules that are effective at blocking quorum sensing. This special report provides an up-to-date review, based on the latest evidence in the literature, of old and new molecules that can positively influence the process of wound healing via their action on MRSA quorum sensing. Quorum sensing-inhibiting molecules, applied topically or injected in situ, have excellent potential to improve both MRSA eradication and quality of wound healing, especially when combined with conventional systemic MRSA therapy. Further human studies are needed to evaluate the efficacy of these molecules.
Collapse
Affiliation(s)
- Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Gianluca Morroni
- Microbiology, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| |
Collapse
|
9
|
Simonetti O, Rizzetto G, Radi G, Molinelli E, Cirioni O, Giacometti A, Offidani A. New Perspectives on Old and New Therapies of Staphylococcal Skin Infections: The Role of Biofilm Targeting in Wound Healing. Antibiotics (Basel) 2021; 10:antibiotics10111377. [PMID: 34827315 PMCID: PMC8615132 DOI: 10.3390/antibiotics10111377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/06/2021] [Accepted: 11/07/2021] [Indexed: 12/31/2022] Open
Abstract
Among the most common complications of both chronic wound and surgical sites are staphylococcal skin infections, which slow down the wound healing process due to various virulence factors, including the ability to produce biofilms. Furthermore, staphylococcal skin infections are often caused by methicillin-resistant Staphylococcus aureus (MRSA) and become a therapeutic challenge. The aim of this narrative review is to collect the latest evidence on old and new anti-staphylococcal therapies, assessing their anti-biofilm properties and their effect on skin wound healing. We considered antibiotics, quorum sensing inhibitors, antimicrobial peptides, topical dressings, and antimicrobial photo-dynamic therapy. According to our review of the literature, targeting of biofilm is an important therapeutic choice in acute and chronic infected skin wounds both to overcome antibiotic resistance and to achieve better wound healing.
Collapse
Affiliation(s)
- Oriana Simonetti
- Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy; (G.R.); (G.R.); (E.M.); (A.O.)
- Correspondence: ; Tel.: +39-0-715-963-494
| | - Giulio Rizzetto
- Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy; (G.R.); (G.R.); (E.M.); (A.O.)
| | - Giulia Radi
- Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy; (G.R.); (G.R.); (E.M.); (A.O.)
| | - Elisa Molinelli
- Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy; (G.R.); (G.R.); (E.M.); (A.O.)
| | - Oscar Cirioni
- Department of Biomedical Sciences and Public Health Clinic of Infectious Diseases, Polytechnic University of Marche, 60020 Ancona, Italy; (O.C.); (A.G.)
| | - Andrea Giacometti
- Department of Biomedical Sciences and Public Health Clinic of Infectious Diseases, Polytechnic University of Marche, 60020 Ancona, Italy; (O.C.); (A.G.)
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy; (G.R.); (G.R.); (E.M.); (A.O.)
| |
Collapse
|
10
|
Ahmad-Mansour N, Loubet P, Pouget C, Dunyach-Remy C, Sotto A, Lavigne JP, Molle V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins (Basel) 2021; 13:677. [PMID: 34678970 PMCID: PMC8540901 DOI: 10.3390/toxins13100677] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus is a clinically important pathogen that causes a wide range of human infections, from minor skin infections to severe tissue infection and sepsis. S. aureus has a high level of antibiotic resistance and is a common cause of infections in hospitals and the community. The rising prevalence of community-acquired methicillin-resistant S. aureus (CA-MRSA), combined with the important severity of S. aureus infections in general, has resulted in the frequent use of anti-staphylococcal antibiotics, leading to increasing resistance rates. Antibiotic-resistant S. aureus continues to be a major health concern, necessitating the development of novel therapeutic strategies. S. aureus uses a wide range of virulence factors, such as toxins, to develop an infection in the host. Recently, anti-virulence treatments that directly or indirectly neutralize S. aureus toxins have showed promise. In this review, we provide an update on toxin pathogenic characteristics, as well as anti-toxin therapeutical strategies.
Collapse
Affiliation(s)
- Nour Ahmad-Mansour
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| | - Paul Loubet
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Cassandra Pouget
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, 30908 Nîmes, France;
| | - Catherine Dunyach-Remy
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Albert Sotto
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Jean-Philippe Lavigne
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| |
Collapse
|
11
|
de Oliveira A, Pinheiro-Hubinger L, Pereira VC, Riboli DFM, Martins KB, Romero LC, da Cunha MDLRDS. Staphylococcal Biofilm on the Surface of Catheters: Electron Microscopy Evaluation of the Inhibition of Biofilm Growth by RNAIII Inhibiting Peptide. Antibiotics (Basel) 2021; 10:antibiotics10070879. [PMID: 34356800 PMCID: PMC8300745 DOI: 10.3390/antibiotics10070879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/04/2022] Open
Abstract
Staphylococcus aureus and coagulase-negative staphylococci (CoNS) have become the main causative agents of medical device-related infections due to their biofilm-forming capability, which protects them from the host’s immune system and from the action of antimicrobials. This study evaluated the ability of RNA III inhibiting peptide (RIP) to inhibit biofilm formation in 10 strains isolated from clinical materials, including one S. aureus strain, two S. epidermidis, two S. haemolyticus, two S. lugdunensis, and one isolate each of the following species: S. warneri, S. hominis, and S. saprophyticus. The isolates were selected from a total of 200 strains evaluated regarding phenotypic biofilm production and the presence and expression of the ica operon. The isolates were cultured in trypticase soy broth with 2% glucose in 96-well polystyrene plates containing catheter segments in the presence and absence of RIP. The catheter segments were observed by scanning electron microscopy. The results showed inhibition of biofilm formation in the presence of RIP in all CoNS isolates; however, RIP did not interfere with biofilm formation by S. aureus. RIP is a promising tool that might be used in the future for the prevention of biofilm-related infections caused by CoNS.
Collapse
Affiliation(s)
- Adilson de Oliveira
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| | - Luiza Pinheiro-Hubinger
- Department of Anatomic Pathology, Lauro de Souza Lima Institute, Bauru 17034-971, Brazil
- Correspondence: ; Tel.: +55-(0)14-38800428
| | - Valéria Cataneli Pereira
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| | - Danilo Flávio Moraes Riboli
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| | - Katheryne Benini Martins
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| | - Letícia Calixto Romero
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| | - Maria de Lourdes Ribeiro de Souza da Cunha
- Department of Chemical and Biological Sciences, Biosciences Institute, UNESP—Universidade Estadual Paulista, Botucatu 18618-691, Brazil; (A.d.O.); (V.C.P.); (D.F.M.R.); (K.B.M.); (L.C.R.); (M.d.L.R.d.S.d.C.)
| |
Collapse
|
12
|
Guzmán-Soto I, McTiernan C, Gonzalez-Gomez M, Ross A, Gupta K, Suuronen EJ, Mah TF, Griffith M, Alarcon EI. Mimicking biofilm formation and development: Recent progress in in vitro and in vivo biofilm models. iScience 2021; 24:102443. [PMID: 34013169 PMCID: PMC8113887 DOI: 10.1016/j.isci.2021.102443] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biofilm formation in living organisms is associated to tissue and implant infections, and it has also been linked to the contribution of antibiotic resistance. Thus, understanding biofilm development and being able to mimic such processes is vital for the successful development of antibiofilm treatments and therapies. Several decades of research have contributed to building the foundation for developing in vitro and in vivo biofilm models. However, no such thing as an "all fit" in vitro or in vivo biofilm models is currently available. In this review, in addition to presenting an updated overview of biofilm formation, we critically revise recent approaches for the improvement of in vitro and in vivo biofilm models.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Christopher McTiernan
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Mayte Gonzalez-Gomez
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Alex Ross
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - Keshav Gupta
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Erik J. Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - May Griffith
- Centre de Recherche Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
- Département d'ophtalmologie, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| |
Collapse
|
13
|
Jiang Y, Geng M, Bai L. Targeting Biofilms Therapy: Current Research Strategies and Development Hurdles. Microorganisms 2020; 8:microorganisms8081222. [PMID: 32796745 PMCID: PMC7465149 DOI: 10.3390/microorganisms8081222] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 01/05/2023] Open
Abstract
Biofilms are aggregate of microorganisms in which cells are frequently embedded within a self-produced matrix of extracellular polymeric substance (EPS) and adhere to each other and/or to a surface. The development of biofilm affords pathogens significantly increased tolerances to antibiotics and antimicrobials. Up to 80% of human bacterial infections are biofilm-associated. Dispersal of biofilms can turn microbial cells into their more vulnerable planktonic phenotype and improve the therapeutic effect of antimicrobials. In this review, we focus on multiple therapeutic strategies that are currently being developed to target important structural and functional characteristics and drug resistance mechanisms of biofilms. We thoroughly discuss the current biofilm targeting strategies from four major aspects—targeting EPS, dispersal molecules, targeting quorum sensing, and targeting dormant cells. We explain each aspect with examples and discuss the main hurdles in the development of biofilm dispersal agents in order to provide a rationale for multi-targeted therapy strategies that target the complicated biofilms. Biofilm dispersal is a promising research direction to treat biofilm-associated infections in the future, and more in vivo experiments should be performed to ensure the efficacy of these therapeutic agents before being used in clinic.
Collapse
|
14
|
Contamination of wounds with fecal bacteria in immuno-suppressed mice. Sci Rep 2020; 10:11494. [PMID: 32661287 PMCID: PMC7359036 DOI: 10.1038/s41598-020-68323-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/15/2020] [Indexed: 12/30/2022] Open
Abstract
Immunocompromised patients are predisposed to chronically infected wounds. Especially ulcers in the dorsal region often experience secondary polymicrobial infections. However, current wound infection models mostly use single-strain bacteria. To mimic clinically occurring infections caused by fecal contamination in immunocompromised/immobile patients, which differ significantly from single-strain infections, the present study aimed at the establishment of a new mouse model using infection by fecal bacteria. Dorsal circular excision wounds in immunosuppressed mice were infected with fecal slurry solution in several dilutions up to 1:8,000. Impact of immunosuppressor, bacterial load and timing on development of wound infections was investigated. Wounds were analyzed by scoring, 3D imaging and swab analyses. Autofluorescence imaging was not successful. Dose-finding of cyclophosphamide-induced immunosuppression was necessary for establishment of bacterial wound infections. Infection with fecal slurry diluted 1:166 to 1:400 induced significantly delayed wound healing (p < 0.05) without systemic reactions. Swab analyses post-infection matched the initial polymicrobial suspension. The customized wound score confirmed significant differences between the groups (p < 0.05). Here we report the establishment of a simple, new mouse model for clinically occurring wound infections by fecal bacteria and the evaluation of appropriate wound analysis methods. In the future, this model will provide a suitable tool for the investigation of complex microbiological interactions and evaluation of new therapeutic approaches.
Collapse
|
15
|
|
16
|
Agostinho Hunt AM, Gibson JA, Larrivee CL, O'Reilly S, Navitskaya S, Needle DB, Abramovitch RB, Busik JV, Waters CM. A bioluminescent Pseudomonas aeruginosa wound model reveals increased mortality of type 1 diabetic mice to biofilm infection. J Wound Care 2019; 26:S24-S33. [PMID: 28704171 DOI: 10.12968/jowc.2017.26.sup7.s24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To examine how bacterial biofilms, as contributing factors in the delayed closure of chronic wounds in patients with diabetes, affect the healing process. METHOD We used daily microscopic imaging and the IVIS Spectrum in vivo imaging system to monitor biofilm infections of bioluminescent Pseudomonas aeruginosa and evaluate healing in non-diabetic and streptozotocin-induced diabetic mice. RESULTS Our studies determined that diabetes alone did not affect the rate of healing of full-depth murine back wounds compared with non-diabetic mice. The application of mature biofilms to the wounds significantly decreased the rate of healing compared with non-infected wounds for both non-diabetic as well as diabetic mice. Diabetic mice were also more severely affected by biofilms displaying elevated pus production, higher mortality rates and statistically significant increase in wound depth, granulation/fibrosis and biofilm presence. Introduction of a mutant Pseudomonas aeruginosa capable of producing high concentrations of cyclic di-GMP did not result in increased persistence in either diabetic or non-diabetic animals compared with the wild type strain. CONCLUSION Understanding the interplay between diabetes and biofilms may lead to novel treatments and better clinical management of chronic wounds.
Collapse
Affiliation(s)
- A M Agostinho Hunt
- Postdoctoral Associate, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI US
| | - J A Gibson
- Undergraduate Researcher, Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI US
| | - C L Larrivee
- Undergraduate Researcher, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI US
| | - S O'Reilly
- Research Assistant Professor, Department of Physiology, Michigan State University, East Lansing, MI USA
| | - S Navitskaya
- Lab Manager, Department of Physiology, Michigan State University, East Lansing, MI USA
| | - D B Needle
- Senior Veterinary Pathologist, New Hampshire Veterinary Diagnostic Laboratory, University of New Hampshire, Durham, NH US
| | - R B Abramovitch
- Assistant Professor, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI US
| | - J V Busik
- Professor, Department of Physiology, Michigan State University, East Lansing, MI USA
| | - C M Waters
- Associate Professor, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI US
| |
Collapse
|
17
|
Multidrug-Resistant Bacteria and Alternative Methods to Control Them: An Overview. Microb Drug Resist 2019; 25:890-908. [DOI: 10.1089/mdr.2018.0319] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
18
|
Enhanced Efficacy of Combinations of Pexiganan with Colistin Versus Acinetobacter Baumannii in Experimental Sepsis. Shock 2018; 46:219-25. [PMID: 26849630 DOI: 10.1097/shk.0000000000000584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We investigated the efficacy of colistin combined with pexiganan in experimental mouse models of Acinetobacter baumannii infection.Adult male BALB/c mice received intraperitoneally 1 mL saline containing 2 × 10 CFU of susceptible and multiresistant A. baumannii. Two hours after bacterial challenge, animals received 1 mg/kg of colistin, 1 mg/kg of pexiganan, or 1 mg/kg of colistin plus 1 mg/kg of pexiganan.Blood culture positivity, the quantities of bacteria in the intra-abdominal fluid, the rate of lethality and immunological studies, such as immunophenotyping and NK cytotoxicity, were evaluated.In the in vitro study, A. baumannii showed susceptibility to colistin and pexiganan and a strong synergy was observed by testing colistin combined with pexiganan with fractionary inhibitory concentration index of 0.312 for both strains.In the in vivo study colistin or pexiganan alone showed a good antimicrobial efficacy. When colistin was combined with pexiganan, the positive interaction produced low bacterial counts that were statistically significant versus singly treated groups. For both strains the highest rate of survival was observed in combined-treated groups (90%).Pexiganan increased NK cytotoxic activity over the levels of infected and colistin-treated animals.In conclusion, pexiganan combined with colistin was found to be efficacious against A. baumannii infection.
Collapse
|
19
|
Koo H, Allan RN, Howlin RP, Hall-Stoodley L, Stoodley P. Targeting microbial biofilms: current and prospective therapeutic strategies. Nat Rev Microbiol 2017; 15:740-755. [PMID: 28944770 PMCID: PMC5685531 DOI: 10.1038/nrmicro.2017.99] [Citation(s) in RCA: 1004] [Impact Index Per Article: 143.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Biofilm formation is a key virulence factor for a wide range of microorganisms that cause chronic infections. The multifactorial nature of biofilm development and drug tolerance imposes great challenges for the use of conventional antimicrobials and indicates the need for multi-targeted or combinatorial therapies. In this Review, we focus on current therapeutic strategies and those under development that target vital structural and functional traits of microbial biofilms and drug tolerance mechanisms, including the extracellular matrix and dormant cells. We emphasize strategies that are supported by in vivo or ex vivo studies, highlight emerging biofilm-targeting technologies and provide a rationale for multi-targeted therapies aimed at disrupting the complex biofilm microenvironment.
Collapse
Affiliation(s)
- Hyun Koo
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, PA, USA
| | - Raymond N Allan
- Clinical and Experimental Sciences, Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
- Southampton NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Robert P Howlin
- Centre for Biological Sciences, University of Southampton, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Luanne Hall-Stoodley
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Department of Microbial Infection and Immunity, Centre for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, USA
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, Centre for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, USA
- Depts. Orthopaedics and Microbiology, The Ohio State University, Columbus, Ohio, USA
- National Center for Advanced Tribology at Southampton (nCATS), Faculty of Engineering and the Environment, University of Southampton, UK
| |
Collapse
|
20
|
Thänert R, Goldmann O, Beineke A, Medina E. Host-inherent variability influences the transcriptional response of Staphylococcus aureus during in vivo infection. Nat Commun 2017; 8:14268. [PMID: 28155859 PMCID: PMC5296661 DOI: 10.1038/ncomms14268] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022] Open
Abstract
The rise of antibiotic resistance calls for alternative strategies to treat bacterial infections. One attractive strategy is to directly target bacterial virulence factors with anti-virulence drugs. The expression of virulence traits by pathogens is, however, not constitutive but rather induced by the level of stress encountered within the host. Here we use dual RNA sequencing (RNA-seq) to show that intrinsic variability in the level of host resistance greatly affects the pathogen's transcriptome in vivo. Through analysis of the transcriptional profiles of host and pathogen during Staphylococcus aureus infection of two mouse strains, shown to be susceptible (A/J) or resistant (C57BL/6) to the pathogen, we demonstrate that the expression of virulence factors is dependent on the encountered host resistance. We furthermore provide evidence that this dependence strongly influences the efficacy of anti-virulence strategies, highlighting a potential limitation for the implementation of these strategies. Drugs inhibiting virulence factors of bacterial pathogens are under development. Here, Thänert et al. analyse the transcriptomes of host and pathogen during Staphylococcus aureus infection of two mouse strains, and show that virulence determinants are differentially expressed in different mouse strains.
Collapse
Affiliation(s)
- Robert Thänert
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Andreas Beineke
- Institute for Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
21
|
Ramasamy M, Lee J. Recent Nanotechnology Approaches for Prevention and Treatment of Biofilm-Associated Infections on Medical Devices. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1851242. [PMID: 27872845 PMCID: PMC5107826 DOI: 10.1155/2016/1851242] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/13/2016] [Indexed: 11/23/2022]
Abstract
Bacterial colonization in the form of biofilms on surfaces causes persistent infections and is an issue of considerable concern to healthcare providers. There is an urgent need for novel antimicrobial or antibiofilm surfaces and biomedical devices that provide protection against biofilm formation and planktonic pathogens, including antibiotic resistant strains. In this context, recent developments in the material science and engineering fields and steady progress in the nanotechnology field have created opportunities to design new biomaterials and surfaces with anti-infective, antifouling, bactericidal, and antibiofilm properties. Here we review a number of the recently developed nanotechnology-based biomaterials and explain underlying strategies used to make antibiofilm surfaces.
Collapse
Affiliation(s)
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
22
|
Simonetti O, Cirioni O, Cacciatore I, Baldassarre L, Orlando F, Pierpaoli E, Lucarini G, Orsetti E, Provinciali M, Fornasari E, Di Stefano A, Giacometti A, Offidani A. Efficacy of the Quorum Sensing Inhibitor FS10 Alone and in Combination with Tigecycline in an Animal Model of Staphylococcal Infected Wound. PLoS One 2016; 11:e0151956. [PMID: 27253706 PMCID: PMC4890846 DOI: 10.1371/journal.pone.0151956] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/06/2016] [Indexed: 11/18/2022] Open
Abstract
In staphylococci, quorum sensing regulates both biofilm formation and toxin production, moreover it has been demonstrated to be inhibited by RNAIII inhibiting peptide (RIP). Aim our study was to evaluate the in vitro activity and its in vivo efficacy of the combined administration of FS10, a novel RIP derivative, and tigecycline in an animal model of methicillin-resistant (MR) and methicillin-sensitive (MS) Staphylococcus aureus wound infection. Using a 1.x2 cm template, one full thickness wound was established through the panniculus carnosus on the back subcutaneous tissue of each animal. Infection was determined by inoculation of 5x107 CFU/ml of bacteria, that produced an abscess within 24 h, after this, treatment was initiated. The study included, for each strain, a control group without infection, a control infected group that did not receive any treatment and a control infected group with drug-free foam dressing, and three infected groups treated, respectively, with: FS10-soaked foam dressing (containing 20 μg FS10), daily intraperitoneal tigecycline (7 mg/Kg), FS10-soaked foam dressing (containing 20 μg FS10) and daily intraperitoneal injections of tigecycline (7 mg/Kg). The main outcome measures were quantitative culture and histological examination of tissue repair. The highest inhibition of infection was achieved in the group that received FS10-soaked and parenteral tigecycline reducing the bacterial load from 107 CFU/ml to about 103 CFU/g for MSSA and to about 104 CFU/g for MRSA. The group treated with FS10-soaked foam dressing associated with parenteral tigecycline showed, histologically, better overall healing with epithelialization and collagen scores significantly higher than those of the other groups in both strains. In conclusion, the combined use of topical FS10 with i.p. tigecycline induced positive interaction in vivo, resulting in an enhanced therapeutic benefit versus staphylococcal infections in murine wound models.
Collapse
Affiliation(s)
- Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences; Università Politecnica delle Marche – Ospedali Riuniti, Ancona, Italy
- * E-mail:
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Italy, Department of Biomedical Sciences and Public Health; Università Politecnica delle Marche – Ospedali Riuniti, Ancona, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara, Italy
| | - Leonardo Baldassarre
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona, Italy
| | - Elisa Pierpaoli
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona, Italy
| | - Guendalina Lucarini
- Clinic of Dermatology, Department of Clinical and Molecular Sciences; Università Politecnica delle Marche, Ancona, Italy
| | - Elena Orsetti
- Clinic of Infectious Diseases, Italy, Department of Biomedical Sciences and Public Health; Università Politecnica delle Marche – Ospedali Riuniti, Ancona, Italy
| | - Mauro Provinciali
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona, Italy
| | - Erika Fornasari
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara, Italy
| | - Antonio Di Stefano
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Italy, Department of Biomedical Sciences and Public Health; Università Politecnica delle Marche – Ospedali Riuniti, Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences; Università Politecnica delle Marche – Ospedali Riuniti, Ancona, Italy
| |
Collapse
|
23
|
Kong C, Neoh HM, Nathan S. Targeting Staphylococcus aureus Toxins: A Potential form of Anti-Virulence Therapy. Toxins (Basel) 2016; 8:toxins8030072. [PMID: 26999200 PMCID: PMC4810217 DOI: 10.3390/toxins8030072] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen and the leading cause of a wide range of severe clinical infections. The range of diseases reflects the diversity of virulence factors produced by this pathogen. To establish an infection in the host, S. aureus expresses an inclusive set of virulence factors such as toxins, enzymes, adhesins, and other surface proteins that allow the pathogen to survive under extreme conditions and are essential for the bacteria’s ability to spread through tissues. Expression and secretion of this array of toxins and enzymes are tightly controlled by a number of regulatory systems. S. aureus is also notorious for its ability to resist the arsenal of currently available antibiotics and dissemination of various multidrug-resistant S. aureus clones limits therapeutic options for a S. aureus infection. Recently, the development of anti-virulence therapeutics that neutralize S. aureus toxins or block the pathways that regulate toxin production has shown potential in thwarting the bacteria’s acquisition of antibiotic resistance. In this review, we provide insights into the regulation of S. aureus toxin production and potential anti-virulence strategies that target S. aureus toxins.
Collapse
Affiliation(s)
- Cin Kong
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| | - Hui-min Neoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
24
|
Abstract
BACKGROUND Infectious complications of musculoskeletal trauma are an important factor contributing to patient morbidity. Biofilm-dispersive bone grafts augmented with D-amino acids (D-AAs) prevent biofilm formation in vitro and in vivo, but the effects of D-AAs on osteocompatibility and new bone formation have not been investigated. QUESTIONS/PURPOSES We asked: (1) Do D-AAs hinder osteoblast and osteoclast differentiation in vitro? (2) Does local delivery of D-AAs from low-viscosity bone grafts inhibit new bone formation in a large-animal model? METHODS Methicillin-sensitive Staphylococcus aureus and methicillin-resistant S aureus clinical isolates, mouse bone marrow stromal cells, and osteoclast precursor cells were treated with an equal mass (1:1:1) mixture of D-Pro:D-Met:D-Phe. The effects of the D-AA dose on biofilm inhibition (n = 4), biofilm dispersion (n = 4), and bone marrow stromal cell proliferation (n = 3) were quantitatively measured by crystal violet staining. Osteoblast differentiation was quantitatively assessed by alkaline phosphatase staining, von Kossa staining, and quantitative reverse transcription for the osteogenic factors a1Col1 and Ocn (n = 3). Osteoclast differentiation was quantitatively measured by tartrate-resistant acid phosphatase staining (n = 3). Bone grafts augmented with 0 or 200 mmol/L D-AAs were injected in ovine femoral condyle defects in four sheep. New bone formation was evaluated by μCT and histology 4 months later. An a priori power analysis indicated that a sample size of four would detect a 7.5% difference of bone volume/total volume between groups assuming a mean and SD of 30% and 5%, respectively, with a power of 80% and an alpha level of 0.05 using a two-tailed t-test between the means of two independent samples. RESULTS Bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation were inhibited at D-AAs concentrations of 27 mmol/L or greater in a dose-responsive manner in vitro (p < 0.05). In methicillin-sensitive and methicillin-resistant S aureus clinical isolates, D-AAs inhibited biofilm formation at concentrations of 13.5 mmol/L or greater in vitro (p < 0.05). Local delivery of D-AAs from low-viscosity grafts did not inhibit new bone formation in a large-animal model pilot study (0 mmol/L D-AAs: bone volume/total volume = 26.9% ± 4.1%; 200 mmol/L D-AAs: bone volume/total volume = 28.3% ± 15.4%; mean difference with 95% CI = -1.4; p = 0.13). CONCLUSIONS D-AAs inhibit biofilm formation, bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation in vitro in a dose-responsive manner. Local delivery of D-AAs from bone grafts did not inhibit new bone formation in vivo at clinically relevant doses. CLINICAL RELEVANCE Local delivery of D-AAs is an effective antibiofilm strategy that does not appear to inhibit bone repair. Longitudinal studies investigating bacterial burden, bone formation, and bone remodeling in contaminated defects as a function of D-AA dose are required to further support the use of D-AAs in the clinical management of infected open fractures.
Collapse
|
25
|
Kaneti G, Meir O, Mor A. Controlling bacterial infections by inhibiting proton-dependent processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:995-1003. [PMID: 26522076 DOI: 10.1016/j.bbamem.2015.10.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
Bacterial resistance to antibiotics is recognized as one of the greatest threats in modern healthcare, taking a staggering toll worldwide. New approaches for controlling bacterial infections must be designed, eventually combining multiple strategies for complimentary therapies. This review explores an old/new paradigm for multi-targeted antibacterial therapy, focused at disturbing bacterial cytoplasmic membrane functions at sub minimal inhibitory concentrations, namely through superficial physical alterations of the bilayer, thereby perturbing transmembrane signals transduction. Such a paradigm may have the advantage of fighting the infection while avoiding many of the known resistance mechanisms. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.
Collapse
Affiliation(s)
- Galoz Kaneti
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ohad Meir
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Amram Mor
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
26
|
Influence of microbiome species in hard-to-heal wounds on disease severity and treatment duration. Braz J Infect Dis 2015; 19:604-13. [PMID: 26518264 PMCID: PMC9425364 DOI: 10.1016/j.bjid.2015.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 08/04/2015] [Accepted: 08/08/2015] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Infections, mostly those associated with colonization of wound by different pathogenic microorganisms, are one of the most serious health complications during a medical treatment. Therefore, this study is focused on the isolation, characterization, and identification of microorganisms prevalent in superficial wounds of patients (n=50) presenting with bacterial infection. METHODS After successful cultivation, bacteria were processed and analyzed. Initially the identification of the strains was performed through matrix-assisted laser desorption/ionization time-of-flight mass spectrometry based on comparison of protein profiles (2-30kDa) with database. Subsequently, bacterial strains from infected wounds were identified by both matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and sequencing of 16S rRNA gene 108. RESULTS The most prevalent species was Staphylococcus aureus (70%), and out of those 11% turned out to be methicillin-resistant (mecA positive). Identified strains were compared with patients' diagnoses using the method of artificial neuronal network to assess the association between severity of infection and wound microbiome species composition. Artificial neuronal network was subsequently used to predict patients' prognosis (n=9) with 85% success. CONCLUSIONS In all of 50 patients tested bacterial infections were identified. Based on the proposed artificial neuronal network we were able to predict the severity of the infection and length of the treatment.
Collapse
|
27
|
Ganesh K, Sinha M, Mathew-Steiner SS, Das A, Roy S, Sen CK. Chronic Wound Biofilm Model. Adv Wound Care (New Rochelle) 2015; 4:382-388. [PMID: 26155380 DOI: 10.1089/wound.2014.0587] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/05/2014] [Indexed: 12/27/2022] Open
Abstract
Significance: Multispecies microbial biofilms may contribute to wound chronicity by derailing the inherent reparative process of the host tissue. In the biofilm form, bacteria are encased within an extracellular polymeric substance and become recalcitrant to antimicrobials and host defenses. For biofilms of relevance to human health, there are two primary contributing factors: the microbial species involved and host response which, in turn, shapes microbial processes over time. This progressive interaction between microbial species and the host is an iterative process that helps evolve an acute-phase infection to a pathogenic chronic biofilm. Thus, long-term wound infection studies are needed to understand the longitudinal cascade of events that culminate into a pathogenic wound biofilm. Recent Advances: Our laboratory has recently published the first long-term (2 month) study of polymicrobial wound biofilm infection in a translationally valuable porcine wound model. Critical Issues: It is widely recognized that the porcine system represents the most translationally valuable approach to experimentally model human skin wounds. A meaningful experimental biofilm model must be in vivo, include mixed species of clinically relevant microbes, and be studied longitudinally long term. Cross-validation of such experimental findings with findings from biofilm-infected patient wounds is critically important. Future Directions: Additional value may be added to the experimental system described above by studying pigs with underlying health complications (e.g., metabolic syndrome), as is typically seen in patient populations.
Collapse
Affiliation(s)
- Kasturi Ganesh
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Mithun Sinha
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Shomita S. Mathew-Steiner
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Amitava Das
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Chandan K. Sen
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| |
Collapse
|
28
|
Biofilm-related infections: bridging the gap between clinical management and fundamental aspects of recalcitrance toward antibiotics. Microbiol Mol Biol Rev 2015; 78:510-43. [PMID: 25184564 DOI: 10.1128/mmbr.00013-14] [Citation(s) in RCA: 784] [Impact Index Per Article: 87.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Surface-associated microbial communities, called biofilms, are present in all environments. Although biofilms play an important positive role in a variety of ecosystems, they also have many negative effects, including biofilm-related infections in medical settings. The ability of pathogenic biofilms to survive in the presence of high concentrations of antibiotics is called "recalcitrance" and is a characteristic property of the biofilm lifestyle, leading to treatment failure and infection recurrence. This review presents our current understanding of the molecular mechanisms of biofilm recalcitrance toward antibiotics and describes how recent progress has improved our capacity to design original and efficient strategies to prevent or eradicate biofilm-related infections.
Collapse
|
29
|
Cooper RA, Bjarnsholt T, Alhede M. Biofilms in wounds: a review of present knowledge. J Wound Care 2015; 23:570, 572-4, 576-80 passim. [PMID: 25375405 DOI: 10.12968/jowc.2014.23.11.570] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Following confirmation of the presence of biofilms in chronic wounds, the term biofilm became a buzzword within the wound healing community. For more than a century pathogens have been successfully isolated and identified from wound specimens using techniques that were devised in the nineteenth century by Louis Pasteur and Robert Koch. Although this approach still provides valuable information with which to help diagnose acute infections and to select appropriate antibiotic therapies, it is evident that those organisms isolated from clinical specimens with the conditions normally used in diagnostic laboratories are mainly in a planktonic form that is unrepresentative of the way in which most microbial species exist naturally. Usually microbial species adhere to each other, as well as to living and non-living surfaces, where they form complex communities surrounded by collectively secreted extracellular polymeric substances (EPS). Cells within such aggregations (or biofilms) display varying physiological and metabolic properties that are distinct from those of planktonic cells, and which contribute to their persistence. There are many factors that influence healing in wounds and the discovery of biofilms in chronic wounds has provided new insight into the reasons why. Increased tolerance of biofilms to antimicrobial agents explains the limited efficacy of antimicrobial agents in chronic wounds and illustrates the need to develop new management strategies. This review aims to explain the nature of biofilms, with a view to explaining their impact on wounds.
Collapse
Affiliation(s)
- R A Cooper
- Professor of Microbiology, Cardiff School of Health Sciences, Cardiff Metropolitan University, Western Avenue, Cardiff, CF5 2YB, S. Wales, UK
| | | | | |
Collapse
|
30
|
Gilbert SR, Camara J, Camara R, Duffy L, Waites K, Kim H, Zinn K. Contaminated open fracture and crush injury: a murine model. Bone Res 2015; 3:14050. [PMID: 26273534 PMCID: PMC4472147 DOI: 10.1038/boneres.2014.50] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/16/2014] [Accepted: 12/27/2014] [Indexed: 12/27/2022] Open
Abstract
Modern warfare has caused a large number of severe extremity injuries, many of which become infected. In more recent conflicts, a pattern of co-infection with Acinetobacter baumannii and methicillin-resistant Staphylococcus aureus has emerged. We attempted to recreate this pattern in an animal model to evaluate the role of vascularity in contaminated open fractures. Historically, it has been observed that infected bones frequently appear hypovascular, but vascularity in association with bone infection has not been examined in animal models. Adult rats underwent femur fracture and muscle crush injury followed by stabilization and bacterial contamination with A. baumannii complex and methicillin-resistant Staphylococcus aureus. Vascularity and perfusion were assessed by microCT angiography and SPECT scanning, respectively, at 1, 2 and 4 weeks after injury. Quantitative bacterial cultures were also obtained. Multi-bacterial infections were successfully created, with methicillin-resistant S. aureus predominating. There was overall increase in blood flow to injured limbs that was markedly greater in bacteria-inoculated limbs. Vessel volume was greater in the infected group. Quadriceps atrophy was seen in both groups, but was greater in the infected group. In this animal model, infected open fractures had greater perfusion and vascularity than non-infected limbs.
Collapse
Affiliation(s)
- Shawn R Gilbert
- Department of Surgery, University of Alabama at Birmingham , AL USA
| | | | | | - Lynn Duffy
- Departments of Pathology, University of Alabama at Birmingham , AL USA
| | - Ken Waites
- Departments of Pathology, University of Alabama at Birmingham , AL USA
| | - Hyunki Kim
- Department of Radiology, University of Alabama at Birmingham , AL USA
| | - Kurt Zinn
- Department of Radiology, University of Alabama at Birmingham , AL USA
| |
Collapse
|
31
|
Brackman G, Coenye T. In Vitro and In Vivo Biofilm Wound Models and Their Application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 897:15-32. [DOI: 10.1007/5584_2015_5002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
32
|
Simonetti O, Cirioni O, Ghiselli R, Orlando F, Silvestri C, Mazzocato S, Kamysz W, Kamysz E, Provinciali M, Giacometti A, Guerrieri M, Offidani A. In vitro activity and in vivo animal model efficacy of IB-367 alone and in combination with imipenem and colistin against Gram-negative bacteria. Peptides 2014; 55:17-22. [PMID: 24531033 DOI: 10.1016/j.peptides.2014.01.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/30/2014] [Accepted: 01/30/2014] [Indexed: 01/22/2023]
Abstract
The aim of our study was to evaluate the in vitro activity of IB-367 and its bactericidal effect for Pseudomonas aeruginosa and Escherichia coli, associated to a synergic study to test the antibiotic combinations between the peptide and colistin or imipenem. Minimum inhibitory concentrations (MICs), the minimum bactericidal concentrations (MBCs), the synergy test and killing study were carried out to evaluate the IB-367 activity. In the in vivo model, a wound was incised through the panniculus carnosus of BALB/c mice, and then inoculated with 5 × 107 colony-forming units of P. aeruginosa and E. coli. For each strain, the study included an infected or not infected group that did not receive any treatment, and five contaminated groups treated with local IB- 367, intraperitoneal imipenem, intraperitoneal colistin, topical IB-367 local plus intraperitoneal imipenem or intraperitoneal colistin. All isolates were inhibited by IB-367 at concentrations of 4-64 mg/l. Killing by IB-367 was shown to be very rapid: its activity on all Gram-negative bacteria was completed within a 40 min exposure period at a concentration of 2 × MIC/l. Synergy was demonstrated when IB-367 was combined with colistin or imipenem. In in vivo studies, the groups treated with topical IB-367 and intraperitoneal colistin showed the best results in terms of bacterial load inhibition either for Pseudomonas or for E. coli. The good in vitro activity and in vivo efficacy, as well as, the synergic interactions with antibiotics suggest that IB-367 is a promising candidate for potential application in the treatment of wound Gram-negative infections.
Collapse
Affiliation(s)
- Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy.
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Roberto Ghiselli
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona, Italy
| | - Carmela Silvestri
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Susanna Mazzocato
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Wojciech Kamysz
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | | | - Mauro Provinciali
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Mario Guerrieri
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| |
Collapse
|
33
|
Simonetti O, Silvestri C, Arzeni D, Cirioni O, Kamysz W, Conte I, Staffolani S, Orsetti E, Morciano A, Castelli P, Scalise A, Kamysz E, Offidani AM, Giacometti A, Barchiesi F. In vitroactivity of the protegrin IB-367 alone and in combination compared with conventional antifungal agents against dermatophytes. Mycoses 2013; 57:233-9. [DOI: 10.1111/myc.12148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 09/11/2013] [Accepted: 09/18/2013] [Indexed: 02/02/2023]
Affiliation(s)
- Oriana Simonetti
- Department of Dermatology; Università Politecnica delle Marche; Ancona Italy
| | - Carmela Silvestri
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Daniela Arzeni
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Oscar Cirioni
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | | | - Irene Conte
- Department of Dermatology; Università Politecnica delle Marche; Ancona Italy
| | - Silvia Staffolani
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Elena Orsetti
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Angela Morciano
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Pamela Castelli
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Alessandro Scalise
- Department of Plastic and Reconstructive Surgery; Università Politecnica delle Marche; Ancona Italy
| | - Elzbieta Kamysz
- Faculty of Pharmacy; Medical University of Gdansk; Gdansk Poland
| | - Anna Maria Offidani
- Department of Dermatology; Università Politecnica delle Marche; Ancona Italy
| | - Andrea Giacometti
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| | - Francesco Barchiesi
- Department of Infectious Diseases and Public Health; Università Politecnica delle Marche; Ancona Italy
| |
Collapse
|
34
|
Simonetti O, Cirioni O, Mocchegiani F, Cacciatore I, Silvestri C, Baldassarre L, Orlando F, Castelli P, Provinciali M, Vivarelli M, Fornasari E, Giacometti A, Offidani A. The efficacy of the quorum sensing inhibitor FS8 and tigecycline in preventing prosthesis biofilm in an animal model of staphylococcal infection. Int J Mol Sci 2013; 14:16321-32. [PMID: 23965956 PMCID: PMC3759913 DOI: 10.3390/ijms140816321] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/01/2013] [Accepted: 07/26/2013] [Indexed: 12/15/2022] Open
Abstract
We investigated the efficacy of tigecycline and FS8, alone or combined, in preventing prosthesis biofilm in a rat model of staphylococcal vascular graft infection. Graft infections were established in the back subcutaneous tissue of adult male Wistar rats by implantation of Dacron prostheses followed by topical inoculation with 2 × 107 colony-forming units of Staphylococcus aureus, strain Smith diffuse. The study included a control group, a contaminated group that did not receive any antibiotic prophylaxis, and three contaminated groups that received: (i) intraperitoneal tigecycline, (ii) FS8-soaked graft, and (iii) tigecycline plus FS8-soaked graft, respectively. Each group included 15 animals. The infection burden was evaluated by using sonication and quantitative agar culture. Moreover, an in vitro binding-study was performed to quantify the how much FS8 was coated to the surface of the prosthesis. Tigecycline, combined with FS8, against the adherent bacteria showed MICs (2.00 mg/L) and MBCs (4.00 mg/L) four-fold lower with respect to tigecycline alone in in vitro studies. The rat groups treated with tigecycline showed the lowest bacterial numbers (4.4 × 104 ± 1.2 × 104 CFU/mL). The FS8-treated group showed a good activity and significant differences compared to control group with bacterial numbers of 6.8 × 104 ± 2.0 × 104 CFU/mL. A stronger inhibition of bacterial growth was observed in rats treated with a combined FS8 and tigecycline therapy than in those that were singly treated with bacterial numbers of 101 CFU/mL graft. In conclusion, the ability to affect biofilm formation as well, its property to be an antibiotic enhancer suggests FS8 as alternative or additional agent to use in conjunction with conventional antimicrobial for prevention of staphylococcal biofilm related infection.
Collapse
Affiliation(s)
- Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-071-5963494; Fax: +39-071-5963446
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Federico Mocchegiani
- Centre for Abdominal Surgery and Organ Transplant, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (F.M.); (M.V.)
| | - Ivana Cacciatore
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Carmela Silvestri
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Leonardo Baldassarre
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona 60100, Italy; E-Mails: (F.O.); (M.P.)
| | - Pamela Castelli
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Mauro Provinciali
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona 60100, Italy; E-Mails: (F.O.); (M.P.)
| | - Marco Vivarelli
- Centre for Abdominal Surgery and Organ Transplant, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (F.M.); (M.V.)
| | - Erika Fornasari
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mail:
| |
Collapse
|
35
|
Sanchez CJ, Prieto EM, Krueger CA, Zienkiewicz KJ, Romano DR, Ward CL, Akers KS, Guelcher SA, Wenke JC. Effects of local delivery of D-amino acids from biofilm-dispersive scaffolds on infection in contaminated rat segmental defects. Biomaterials 2013; 34:7533-43. [PMID: 23831189 DOI: 10.1016/j.biomaterials.2013.06.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/14/2013] [Indexed: 01/10/2023]
Abstract
Infectious complications of open fractures continue to be a significant factor contributing to non-osseous union and extremity amputation. The persistence of bacteria within biofilms despite meticulous debridement and antibiotic therapy is believed to be a major cause of chronic infection. Considering the difficulties in treating biofilm-associated infections, the use of biofilm dispersal agents as a therapeutic strategy for the prevention of biofilm-associated infections has gained considerable interest. In this study, we investigated whether local delivery of D-Amino Acids (D-AAs), a biofilm dispersal agent, protects scaffolds from contamination and reduces microbial burden within contaminated rat segmental defects in vivo. In vitro testing on biofilms of clinical isolates of Staphylococcus aureus demonstrated that D-Met, D-Phe, D-Pro, and D-Trp were highly effective at dispersing and preventing biofilm formation individually, and the effect was enhanced for an equimolar mixture of D-AAs. Incorporation of D-AAs into polyurethane scaffolds as a mixture (1:1:1 D-Met:D-Pro:D-Trp) significantly reduced bacterial contamination on the scaffold surface in vitro and within bone when implanted into contaminated femoral segmental defects. Our results underscore the potential of local delivery of d-AAs for reducing bacterial contamination by targeting bacteria within biofilms, which may represent a treatment strategy for improving healing outcomes associated with open fractures.
Collapse
Affiliation(s)
- Carlos J Sanchez
- United States Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine Task Area, Fort Sam Houston, San Antonio, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lebeaux D, Chauhan A, Rendueles O, Beloin C. From in vitro to in vivo Models of Bacterial Biofilm-Related Infections. Pathogens 2013; 2:288-356. [PMID: 25437038 PMCID: PMC4235718 DOI: 10.3390/pathogens2020288] [Citation(s) in RCA: 308] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/01/2013] [Accepted: 05/08/2013] [Indexed: 12/13/2022] Open
Abstract
The influence of microorganisms growing as sessile communities in a large number of human infections has been extensively studied and recognized for 30–40 years, therefore warranting intense scientific and medical research. Nonetheless, mimicking the biofilm-life style of bacteria and biofilm-related infections has been an arduous task. Models used to study biofilms range from simple in vitro to complex in vivo models of tissues or device-related infections. These different models have progressively contributed to the current knowledge of biofilm physiology within the host context. While far from a complete understanding of the multiple elements controlling the dynamic interactions between the host and biofilms, we are nowadays witnessing the emergence of promising preventive or curative strategies to fight biofilm-related infections. This review undertakes a comprehensive analysis of the literature from a historic perspective commenting on the contribution of the different models and discussing future venues and new approaches that can be merged with more traditional techniques in order to model biofilm-infections and efficiently fight them.
Collapse
Affiliation(s)
- David Lebeaux
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Ashwini Chauhan
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Olaya Rendueles
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Christophe Beloin
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| |
Collapse
|
37
|
|
38
|
Cirioni O, Mocchegiani F, Cacciatore I, Vecchiet J, Silvestri C, Baldassarre L, Ucciferri C, Orsetti E, Castelli P, Provinciali M, Vivarelli M, Fornasari E, Giacometti A. Quorum sensing inhibitor FS3-coated vascular graft enhances daptomycin efficacy in a rat model of staphylococcal infection. Peptides 2013; 40:77-81. [PMID: 23262356 DOI: 10.1016/j.peptides.2012.12.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/04/2012] [Accepted: 12/04/2012] [Indexed: 01/01/2023]
Abstract
The aim of the study was to investigate the efficacy of the quorum sensing inhibitor FS3 and daptomycin in preventing prosthesis biofilm in a rat model of staphylococcal vascular graft infection. Graft infections were established in the back subcutaneous tissue of adult male Wistar rats by implantation of Dacron prostheses followed by topical inoculation with 2×10(7) colony-forming units of Staphylococcus aureus, strain Smith diffuse. The study included a control group, a contaminated group that did not receive any antibiotic prophylaxis and three contaminated groups that received: (i) intraperitoneal daptomycin, (ii) FS3-soacked graft, and (iii) daptomycin plus FS3-soaked graft, respectively. Each group included 15 animals. The infection burden was evaluated by using sonication and quantitative agar culture. Moreover, an in vitro binding-study was performed to quantify the how much FS3 was coated to the surface of the prosthesis. The in vitro studies showed, that minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values for daptomycin were lower in presence of FS3. In in vivo studies, when tested alone, daptomycin and FS3 showed good efficacies. Their combination showed efficacies significantly higher than that of each single compound. Daptomycin is an important candidate for prevention of staphylococcal biofilm related infection and FS3 could serve as an interesting anti-staphylococcal antibiotic enhancer.
Collapse
Affiliation(s)
- Oscar Cirioni
- Clinic of Infectious Diseases, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
In vivo modeling of biofilm-infected wounds: A review. J Surg Res 2012; 178:330-8. [DOI: 10.1016/j.jss.2012.06.048] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/17/2012] [Accepted: 06/21/2012] [Indexed: 12/23/2022]
|
40
|
Fitzgerald-Hughes D, Devocelle M, Humphreys H. Beyond conventional antibiotics for the future treatment of methicillin-resistantStaphylococcus aureusinfections: two novel alternatives. ACTA ACUST UNITED AC 2012; 65:399-412. [DOI: 10.1111/j.1574-695x.2012.00954.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 03/02/2012] [Accepted: 03/02/2012] [Indexed: 12/31/2022]
|
41
|
Silvestri C, Cirioni O, Arzeni D, Ghiselli R, Simonetti O, Orlando F, Ganzetti G, Staffolani S, Brescini L, Provinciali M, Offidani A, Guerrieri M, Giacometti A. In vitro activity and in vivo efficacy of tigecycline alone and in combination with daptomycin and rifampin against Gram-positive cocci isolated from surgical wound infection. Eur J Clin Microbiol Infect Dis 2011; 31:1759-64. [DOI: 10.1007/s10096-011-1498-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 11/16/2011] [Indexed: 12/30/2022]
|
42
|
Provinciali M, Cirioni O, Orlando F, Pierpaoli E, Barucca A, Silvestri C, Ghiselli R, Scalise A, Brescini L, Guerrieri M, Giacometti A. Vitamin E improves the in vivo efficacy of tigecycline and daptomycin in an animal model of wounds infected with meticillin-resistant Staphylococcus aureus. J Med Microbiol 2011; 60:1806-1812. [DOI: 10.1099/jmm.0.032516-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Mauro Provinciali
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA-IRCCS, Università Politecnica delle Marche, Ancona, Italy
| | - Oscar Cirioni
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA-IRCCS, Università Politecnica delle Marche, Ancona, Italy
| | - Elisa Pierpaoli
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA-IRCCS, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandra Barucca
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA-IRCCS, Università Politecnica delle Marche, Ancona, Italy
| | - Carmela Silvestri
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Roberto Ghiselli
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandro Scalise
- Department of Plastic and Reconstructive Surgery General Surgery, Università Politecnica delle Marche, Ancona, Italy
| | - Lucia Brescini
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Mario Guerrieri
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Giacometti
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
43
|
Bharathan M, Mullarky IK. Targeting mucosal immunity in the battle to develop a mastitis vaccine. J Mammary Gland Biol Neoplasia 2011; 16:409-19. [PMID: 21968537 DOI: 10.1007/s10911-011-9233-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/14/2011] [Indexed: 01/31/2023] Open
Abstract
The mucosal immune system encounters antigens that enhance and suppress immune function, and serves as a selective barrier against invading pathogens. The mammary gland not only encounters antigens but also produces a nutrient evolved to protect and enhance mucosal development in the neonate. Efforts to manipulate antibody concentrations in milk to prevent mastitis, an infection of the mammary gland, have been hampered both by complexity and variation in target pathogens and limited knowledge of cellular immunity in the gland. Successful vaccination strategies must overcome the natural processes that regulate types and concentrations of milk antibodies for neonatal development, and enhance cellular immunity. Furthermore, the need to overcome dampening of immunity caused by non-pathogenic encounters to successfully prevent establishment of infection is an additional obstacle in vaccine development at mucosal sites. A significant mastitis pathogen, Staphylococcus aureus, not only resides as a normal flora on a multitude of species, but also causes clinical disease with limited treatment options. Using the bovine model of S. aureus mastitis, researchers can decipher the role of antigen selection and presentation by mammary dendritic cells, enhance development of central and effector memory function, and subsequently target specific memory cells to the mammary gland for successful vaccine development. This brief review provides an overview of adaptive immunity, previous vaccine efforts, current immunological findings relevant to enhancing immune memory, and research technologies that show promise in directing future vaccine efforts to enhance mammary gland immunity and prevent mastitis.
Collapse
Affiliation(s)
- Mini Bharathan
- Immunology, Human Therapeutic Division, Intrexon Corporation, Germantown, MD, USA
| | | |
Collapse
|
44
|
Simonetti O, Cirioni O, Lucarini G, Orlando F, Ghiselli R, Silvestri C, Brescini L, Rocchi M, Provinciali M, Guerrieri M, Di Primio R, Giacometti A, Offidani A. Tigecycline accelerates staphylococcal-infected burn wound healing through matrix metalloproteinase-9 modulation. J Antimicrob Chemother 2011; 67:191-201. [PMID: 22065244 DOI: 10.1093/jac/dkr440] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES We investigated the in vivo efficacy of tigecycline, a new glycylcycline (a tetracycline derivative), in the management of methicillin-resistant Staphylococcus aureus (MRSA)-infected experimental surgical wounds in rats. The main outcome measures were quantitative bacterial culture, histological examination and immunohistochemical expression of matrix metalloproteinase-9 (MMP-9) and collagen IV. METHODS An animal model was used to compare the in vivo efficacy of teicoplanin and tigecycline in the treatment of burn wound infections by S. aureus. A copper bar, heated in boiling water, was placed on the paraspinal site of each rat, resulting in full thickness burns. A small gauze was placed over each burn and then inoculated with 5 × 10(7) cfu of S. aureus ATCC 43300. To mimic the clinical situation in burn patients, surgical debridement was performed 48 h after the injury. The wounds were left to heal by secondary intention. The study included an uninfected control group that did not receive any treatment, a contaminated group that did not receive any treatment, and two contaminated groups treated with intraperitoneal tigecycline (2 mg/kg) and teicoplanin (7 mg/kg), respectively. RESULTS All antibiotic treatments were significantly effective. Tigecycline showed the highest antimicrobial activity, with a better impact on histological results. Infected rats treated with tigecycline showed a significant decrease in MMP-9 expression both in epithelium and in dermis compared with rats treated with teicoplanin. CONCLUSIONS Tigecycline, besides its antimicrobial activity, exerts an important modulatory effect on MMP-9, accelerating wound healing in staphylococcal-infected burns.
Collapse
Affiliation(s)
- Oriana Simonetti
- Department of Dermatology, Università Politecnica delle Marche, Ancona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Leitner G, Krifucks O, Kiran MD, Balaban N. Vaccine development for the prevention of staphylococcal mastitis in dairy cows. Vet Immunol Immunopathol 2011; 142:25-35. [DOI: 10.1016/j.vetimm.2011.03.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 10/18/2022]
|
46
|
Dai T, Kharkwal GB, Tanaka M, Huang YY, Bil de Arce VJ, Hamblin MR. Animal models of external traumatic wound infections. Virulence 2011; 2:296-315. [PMID: 21701256 DOI: 10.4161/viru.2.4.16840] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Despite advances in traumatic wound care and management, infections remain a leading cause of mortality,morbidity and economic disruption in millions of wound patients around the world. Animal models have become standard tools for studying a wide array of external traumatic wound infections and testing new antimicrobial strategies. RESULTS Animal models of external traumatic wound infections reported by different investigators vary in animal species used, microorganism strains, the number of microorganisms applied, the size of the wounds and for burn infections, the length of time the heated object or liquid is in contact with the skin. METHODS This review covers experimental infections in animal models of surgical wounds, skin abrasions, burns, lacerations,excisional wounds and open fractures. CONCLUSIONS As antibiotic resistance continues to increase,more new antimicrobial approaches are urgently needed.These should be tested using standard protocols for infections in external traumatic wounds in animal models.
Collapse
Affiliation(s)
- Tianhong Dai
- Wellman Center for Photomedicine, Massachusetts General Hospital, USA
| | | | | | | | | | | |
Collapse
|
47
|
Cirioni O, Myszka H, Dawgul M, Ghiselli R, Orlando F, Silvestri C, Brescini L, Kamysz W, Guerrieri M, Giacometti A. In vitro activity and in vivo efficacy of the saponin diosgenyl 2-amino-2-deoxy-β-D-glucopyranoside hydrochloride (HSM1) alone and in combination with daptomycin and vancomycin against Gram-positive cocci. J Med Microbiol 2011; 60:1337-1343. [PMID: 21511888 DOI: 10.1099/jmm.0.031708-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Surgical site infections are the second most common hospital- and community-acquired Gram-positive infections, with the US Centers for Disease Control and Prevention estimating that about 500 000 surgical site infections occur annually in the USA. The aim of this work was to determine the in vitro activity of the saponin diosgenyl 2-amino-2-deoxy-β-d-glucopyranoside hydrochloride (HSM1) and its bactericidal effect for a large number of Gram-positive cocci, as well as to investigate its in vitro interaction with seven clinically used antibiotics. In vivo, a wound model was established through the panniculus carnosus of BALB/c mice and then inoculated with 5×10(7) c.f.u. Staphylococcus aureus or Enterococcus faecalis. For each bacterial strain, the study included an infected or non-infected group that did not receive any treatment, a group treated with local HSM1, a group treated with intraperitoneal vancomycin, a group treated with intraperitoneal daptomycin and two groups that received HSM1 local treatment plus intraperitoneal vancomycin or daptomycin. All isolates were inhibited by HSM1 at concentrations of 2-32 mg l(-1). Synergy was demonstrated when HSM1 was combined with vancomycin and daptomycin. In in vivo studies, all groups treated with single drugs showed a statistically significant result compared with the control group. The two groups treated with drug combinations showed the highest antimicrobial efficacy. The good in vitro activities and the in vivo efficacy suggest HSM1 as a promising therapeutic candidate in Gram-positive wound infections.
Collapse
Affiliation(s)
- Oscar Cirioni
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Henryk Myszka
- Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | | | - Roberto Ghiselli
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Units, Research Department, INRCA IRRCS, Ancona, Italy
| | - Carmela Silvestri
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Lucia Brescini
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Wojciech Kamysz
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Mario Guerrieri
- General Surgery and Surgery Methodology Clinic, Università Politecnica delle Marche - Ospedali Riuniti, Ancona, Italy
| | - Andrea Giacometti
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
48
|
Simonetti O, Cirioni O, Orlando F, Alongi C, Lucarini G, Silvestri C, Zizzi A, Fantetti L, Roncucci G, Giacometti A, Offidani A, Provinciali M. Effectiveness of antimicrobial photodynamic therapy with a single treatment of RLP068/Cl in an experimental model of Staphylococcus aureus wound infection. Br J Dermatol 2011; 164:987-95. [PMID: 21275941 DOI: 10.1111/j.1365-2133.2011.10232.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Chronic leg ulceration is a common health problem. It is well known that a clinically relevant bacterial load in chronic cutaneous wounds interferes significantly with the normal process of healing. Staphylococcus aureus is the most important representative of the staphylococcal group which causes clinically relevant infections within immunocompetent patients. OBJECTIVES To investigate the efficacy of a single treatment of antimicrobial photodynamic therapy (APDT) with RLP068/Cl in a mouse model of a surgical wound infection induced with a methicillin-resistant strain of S. aureus (MRSA). METHODS Wounds, established through the panniculus carnosus of BALB/c and CD1 mice, were inoculated with 5 x 10(7) c.f.u. of MRSA. Mice were randomized into four groups respectively receiving no treatment, APDT with placebo, APDT with a new phthalocyanine derivative (RLP068/Cl) and intraperitoneal teicoplanin. RESULTS On day 2 from infection, a strong reduction of bacterial counts (≈ 3 logs) was observed in mice treated with RLP068/Cl in comparison with infected untreated mice. On day 9 from infection, a comparable and significant (≈ 2 logs) reduction of bacterial counts was found in mice treated with RLP068/Cl or with teicoplanin. At this time, histological examinations revealed that wounds treated with RLP068/Cl showed a complete re-epithelialization with a continuous epithelial lining. CONCLUSIONS The results of the in vivo study demonstrated that APDT with RLP068/Cl may be useful in the management of chronic infected wounds, accelerating the repair process through a significant bacterial inhibition.
Collapse
Affiliation(s)
- O Simonetti
- Dermatological Clinic, Department of Clinical Medicine and Applied Biotechnology, Università Politecnica delle Marche, Ancona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lowery CA, Salzameda NT, Sawada D, Kaufmann GF, Janda KD. Medicinal chemistry as a conduit for the modulation of quorum sensing. J Med Chem 2010; 53:7467-89. [PMID: 20669927 DOI: 10.1021/jm901742e] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Colin A Lowery
- Department of Chemistry and Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
50
|
Coenye T, Nelis HJ. In vitro and in vivo model systems to study microbial biofilm formation. J Microbiol Methods 2010; 83:89-105. [DOI: 10.1016/j.mimet.2010.08.018] [Citation(s) in RCA: 251] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 08/19/2010] [Accepted: 08/23/2010] [Indexed: 12/23/2022]
|