1
|
Alanazi HH, Elasbali AM, Alanazi MK, El Azab EF. Medicinal Herbs: Promising Immunomodulators for the Treatment of Infectious Diseases. Molecules 2023; 28:8045. [PMID: 38138535 PMCID: PMC10745476 DOI: 10.3390/molecules28248045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/24/2023] Open
Abstract
Humans are constantly at high risk of emerging pandemics caused by viral and bacterial infections. The emergence of new pandemics is mainly caused by evolved viruses and bacteria that are highly resistant to existing medications. The rapid evolution of infectious agents demands the urgent investigation of new therapeutic strategies to prevent and treat these infections at an early stage. One of these therapeutic strategies includes the use of medicinal herbs for their antibacterial and antiviral properties. The use of herbal medicines as remedies is very ancient and has been employed for centuries. Many studies have confirmed the antimicrobial activities of herbs against various pathogens in vitro and in vivo. The therapeutic effect of medicinal herbs is mainly attributed to the natural bioactive molecules present in these plants such as alkaloids, flavonoids, and terpenoids. Different mechanisms have been proposed for how medicinal herbs enhance the immune system and combat pathogens. Such mechanisms include the disruption of bacterial cell membranes, suppression of protein synthesis, and limitation of pathogen replication through the inhibition of nucleic acid synthesis. Medicinal herbs have been shown to treat a number of infectious diseases by modulating the immune system's components. For instance, many medicinal herbs alleviate inflammation by reducing pro-inflammatory cytokines (e.g., tumor necrosis factor-alpha (TNF-α), interleukin-1, IL-6) while promoting the production of anti-inflammatory cytokines (e.g., IL-10). Medicinal herbs also play a role in defense against viral and intracellular infections by enhancing the proliferation and functions of natural killer cells, T-helper-1 cells, and macrophages. In this review, we will explore the use of the most common herbs in preventing and treating infectious and non-infectious diseases. Using current and recently published studies, we focus on the immunomodulatory and therapeutic effects induced by medicinal herbs to enhance immune responses during diseases.
Collapse
Affiliation(s)
- Hamad H. Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Al-Qurayyat 77455, Saudi Arabia; (A.M.E.); (E.F.E.A.)
| | | | | | | |
Collapse
|
2
|
Prathapan P. A determination of pan-pathogen antimicrobials? MEDICINE IN DRUG DISCOVERY 2022; 14:100120. [PMID: 35098103 PMCID: PMC8785259 DOI: 10.1016/j.medidd.2022.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/01/2022] [Accepted: 01/17/2022] [Indexed: 11/29/2022] Open
Abstract
While antimicrobial drug development has historically mitigated infectious diseases that are known, COVID-19 revealed a dearth of 'in-advance' therapeutics suitable for infections by pathogens that have not yet emerged. Such drugs must exhibit a property that is antithetical to the classical paradigm of antimicrobial development: the ability to treat infections by any pathogen. Characterisation of such 'pan-pathogen' antimicrobials requires consolidation of drug repositioning studies, a new and growing field of drug discovery. In this review, a previously-established system for evaluating repositioning studies is used to highlight 4 therapeutics which exhibit pan-pathogen properties, namely azithromycin, ivermectin, niclosamide, and nitazoxanide. Recognition of the pan-pathogen nature of these antimicrobials is the cornerstone of a novel paradigm of antimicrobial development that is not only anticipatory of pandemics and bioterrorist attacks, but cognisant of conserved anti-infective mechanisms within the host-pathogen interactome which are only now beginning to emerge. Ultimately, the discovery of pan-pathogen antimicrobials is concomitantly the discovery of a new class of antivirals, and begets significant implications for pandemic preparedness research in a world after COVID-19.
Collapse
Affiliation(s)
- Praveen Prathapan
- New Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
3
|
Nagaraj S, Manivannan S, Narayan S. Potent antifungal agents and use of nanocarriers to improve delivery to the infected site: A systematic review. J Basic Microbiol 2021; 61:849-873. [PMID: 34351655 DOI: 10.1002/jobm.202100204] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/29/2021] [Accepted: 08/01/2021] [Indexed: 01/30/2023]
Abstract
There are four major classes of antifungals with the predominant mechanism of action being targeting of cell wall or cell membrane. As in other drugs, low solubility of these compounds has led to low bioavailability in target tissues. Enhanced drug dosages have effects such as toxicity, drug-drug interactions, and increased drug resistance by fungi. This article reviews the current state-of-the-art of antifungals, structure, mechanism of action, other usages, and toxic side effects. The emergence of nanoformulations to transport and uniformly release cargo at the target site is a boon in antifungal treatment. The article details research that lead to the development of nanoformulations of antifungals and potential advantages and avoidance of the lacunae characterizing conventional drugs. A range of nanoformulations based on liposomes, polymers are in various stages of research and their potential advantages have been brought out. It could be observed that under similar dosages, test models, and duration, nanoformulations provided enhanced activity, reduced toxicity, higher uptake and higher immunostimulatory effects. In most instances, the mechanism of antifungal activity of nanoformulations was similar to that of regular antifungal. There are possibilities of coupling multiple antifungals on the same nano-platform. Increased activity coupled with multiple mechanisms of action presents for nanoformulations a tremendous opportunity to overcome antifungal resistance. In the years to come, robust methods for the preparation of nanoformulations taking into account the repeatability and reproducibility in action, furthering the studies on nanoformulation toxicity and studies of human models are required before extensive use of nanoformulations as a prescribed drug.
Collapse
Affiliation(s)
- Saraswathi Nagaraj
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| | - Sivakami Manivannan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| | - Shoba Narayan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| |
Collapse
|
4
|
Vansarla G, Håkansson AP, Bergenfelz C. HAMLET a human milk protein-lipid complex induces a pro-inflammatory phenotype of myeloid cells. Eur J Immunol 2021; 51:965-977. [PMID: 33348422 PMCID: PMC8248127 DOI: 10.1002/eji.202048813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/06/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
HAMLET is a protein‐lipid complex with a specific and broad bactericidal and tumoricidal activity, that lacks cytotoxic activity against healthy cells. In this study, we show that HAMLET also has general immune‐stimulatory effects on primary human monocyte‐derived dendritic cells and macrophages (Mo‐DC and Mo‐M) and murine RAW264.7 macrophages. HAMLET, but not its components alpha‐lactalbumin or oleic acid, induces mature CD14low/–CD83+ Mo‐DC and M1‐like CD14+CD86++ Mo‐M surface phenotypes. Concomitantly, inflammatory mediators, including IL‐2, IL‐6, IL‐10, IL‐12 and MIP‐1α, were released in the supernatant of HAMLET‐stimulated cells, indicating a mainly pro‐inflammatory phenotype. The HAMLET‐induced phenotype was mediated by calcium, NFκB and p38 MAPK signaling in Mo‐DCs and calcium, NFκB and ERK signaling in Mo‐M as inhibitors of these pathways almost completely blocked the induction of mature Mo‐DCs and M1‐like Mo‐M. Compared to unstimulated Mo‐DCs, HAMLET‐stimulated Mo‐DCs were more potent in inducing T cell proliferation and HAMLET‐stimulated macrophages were more efficient in phagocytosis of Streptococcus pneumoniae in vitro. This indicates a functionally activated phenotype of HAMLET‐stimulated DCs and macrophages. Combined, we propose that HAMLET has a two‐fold anti‐bacterial activity; one inducing direct cytotoxic activity, the other indirectly mediating elimination of bacteria by activation of immune cells of the myeloid lineage.
Collapse
Affiliation(s)
- Goutham Vansarla
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, SE-214 28, Sweden
| | - Anders P Håkansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, SE-214 28, Sweden
| | - Caroline Bergenfelz
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, SE-214 28, Sweden
| |
Collapse
|
5
|
|
6
|
Rabbit’s Host Defense Peptide (RSRAH) Protects Mice from Escherichia coli Challenge. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9694-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
7
|
He X, Xiong LH, Zhao Z, Wang Z, Luo L, Lam JWY, Kwok RTK, Tang BZ. AIE-based theranostic systems for detection and killing of pathogens. Theranostics 2019; 9:3223-3248. [PMID: 31244951 PMCID: PMC6567968 DOI: 10.7150/thno.31844] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
Pathogenic bacteria, fungi and viruses pose serious threats to the human health under appropriate conditions. There are many rapid and sensitive approaches have been developed for identification and quantification of specific pathogens, but many challenges still exist. Culture/colony counting and polymerase chain reaction are the classical methods used for pathogen detection, but their operations are time-consuming and laborious. On the other hand, the emergence and rapid spread of multidrug-resistant pathogens is another global threat. It is thus of utmost urgency to develop new therapeutic agents or strategies. Luminogens with aggregation-induced emission (AIEgens) and their derived supramolecular systems with unique optical properties have been developed as fluorescent probes for turn-on sensing of pathogens with high sensitivity and specificity. In addition, AIE-based supramolecular nanostructures exhibit excellent photodynamic inactivation (PDI) activity in aggregate, offering great potential for not only light-up diagnosis of pathogen, but also image-guided PDI therapy for pathogenic infection.
Collapse
Affiliation(s)
- Xuewen He
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Ling-Hong Xiong
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zheng Zhao
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Zaiyu Wang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jacky Wing Yip Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Ryan Tsz Kin Kwok
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Institute for Advanced Study and Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
- NSFC Center for Luminescence from Molecular Aggregates, SCUT-HKUST Joint Research Laboratory, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
8
|
Martinez MN, Watts JL, Gilbert JM. Questions associated with the development of novel drugs intended for the treatment of bacterial infections in veterinary species. Vet J 2019; 248:79-85. [PMID: 31113568 DOI: 10.1016/j.tvjl.2019.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 11/17/2022]
Abstract
The emergence of multi-drug resistant bacteria has limited therapeutic options for the treatment of bacterial diseases in both human and veterinary medicine. This has resulted in an urgent need for novel agents to treat infectious diseases. Veterinary medicine is further constrained by the need to ensure that our emerging therapeutics have minimal or no impact on resistance in human pathogens. Thus, there has recently been increased attention given to the development of alternative treatments for infectious disease in animals. The domain of alternative therapies, which includes antimicrobial peptides, bacteriophages, probiotics, and immunomodulators, provides a means to directly inhibit the ability of a pathogen to damage the host while optimally, not imposing a selective pressure favouring antibiotic resistance. However, it is recognized that bacterial pathogens have the capability of expressing a variety of virulence factors, necessitating a clear understanding of the specific target for that therapeutic intervention. This manuscript explores the various virulence mechanisms, the potential utility of developing novel anti-virulence agents for counteracting the expression of diseases associated with veterinary species, and some of the unique regulatory hurdles to be addressed within the framework of a new animal drug application. We conclude with the public health concerns to be considered as these agents are integrated into the veterinary therapeutic arsenal. Our hope is that this manuscript will provide a platform to stimulate discussions on the critical questions that need to be addressed.
Collapse
Affiliation(s)
- Marilyn N Martinez
- US FDA Center for Veterinary Medicine, Rockville, MD 20855, United States.
| | - Jeffrey L Watts
- Zoetis, Inc., 333 Portage Street, Kalamazoo, MI 49007, United States
| | - Jeffrey M Gilbert
- US FDA Center for Veterinary Medicine, Rockville, MD 20855, United States
| |
Collapse
|
9
|
Abstract
Bacterial infections have been traditionally controlled by antibiotics and vaccines, and these approaches have greatly improved health and longevity. However, multiple stakeholders are declaring that the lack of new interventions is putting our ability to prevent and treat bacterial infections at risk. Vaccine and antibiotic approaches still have the potential to address this threat. Innovative vaccine technologies, such as reverse vaccinology, novel adjuvants, and rationally designed bacterial outer membrane vesicles, together with progress in polysaccharide conjugation and antigen design, have the potential to boost the development of vaccines targeting several classes of multidrug-resistant bacteria. Furthermore, new approaches to deliver small-molecule antibacterials into bacteria, such as hijacking active uptake pathways and potentiator approaches, along with a focus on alternative modalities, such as targeting host factors, blocking bacterial virulence factors, monoclonal antibodies, and microbiome interventions, all have potential. Both vaccines and antibacterial approaches are needed to tackle the global challenge of antimicrobial resistance (AMR), and both areas have the underpinning science to address this need. However, a concerted research agenda and rethinking of the value society puts on interventions that save lives, by preventing or treating life-threatening bacterial infections, are needed to bring these ideas to fruition.
Collapse
|
10
|
Rajasekaran G, Kim EY, Shin SY. LL-37-derived membrane-active FK-13 analogs possessing cell selectivity, anti-biofilm activity and synergy with chloramphenicol and anti-inflammatory activity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:722-733. [PMID: 28161291 DOI: 10.1016/j.bbamem.2017.01.037] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/12/2017] [Accepted: 01/31/2017] [Indexed: 12/21/2022]
Abstract
Although the human-derived antimicrobial peptide (AMP) LL-37 has potent antimicrobial and anti-inflammatory activities, its therapeutic application is limited by its low cell selectivity and high production cost due to its large size. To overcome these problems, we tried to develop novel LL-37-derived short α-helical AMPs with improved cell selectivity and without a significant loss of anti-inflammatory activity relative to that of parental LL-37. Using amino acid substitution, we designed and synthesized a series of FK13 analogs based on the sequence of the 13-meric short FK13 peptide (residues 17-29 of LL-37) that has been identified as the region responsible for the antimicrobial activity of LL-37. Among the designed FK13 analogs, FK-13-a1 and FK-13-a7 showed high cell selectivity and retained the anti-inflammatory activity. The therapeutic index (a measure of cell selectivity) of FK-13-a1 and FK-13-a7 was 6.3- and 2.3-fold that of parental LL-37, respectively. Furthermore, FK-13-a1 and FK-13-a7 displayed more potent antimicrobial activity against antibiotic-resistant bacteria including MRSA, MDRPA, and VREF, than did LL-37. In addition, FK-13-a1 and FK-13-a7 exhibited greater synergistic effects with chloramphenicol against MRSA and MDRPA and were more effective anti-biofilm agents against MDRPA than LL-37 was. Moreover, FK-13-a1 and FK-13-a7 maintained their activities in the presence of physiological salts and human serum. SYTOX green uptake, membrane depolarization and killing kinetics revealed that FK13-a1 and FK13-a7 kills microbial cells by permeabilizing the cell membrane and damaging membrane integrity. Taken together, our results suggest that FK13-a1 and FK13-a7 can be developed as novel antimicrobial/anti-inflammatory agents.
Collapse
Affiliation(s)
- Ganesan Rajasekaran
- Department of Medical Science, Graduate School, Chosun University, Gwangju 501-759, Republic of Korea
| | - Eun Young Kim
- Department of Medical Science, Graduate School, Chosun University, Gwangju 501-759, Republic of Korea
| | - Song Yub Shin
- Department of Medical Science, Graduate School, Chosun University, Gwangju 501-759, Republic of Korea; Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea.
| |
Collapse
|
11
|
PPAR γ in Bacterial Infections: A Friend or Foe? PPAR Res 2016; 2016:7963540. [PMID: 27774097 PMCID: PMC5059608 DOI: 10.1155/2016/7963540] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/21/2016] [Indexed: 01/09/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is now recognized as an important modulator of leukocyte inflammatory responses and function. Its immunoregulatory function has been studied in a variety of contexts, including bacterial infections of the lungs and central nervous system, sepsis, and conditions such as chronic granulomatous disease. Although it is generally believed that PPARγ activation is beneficial for the host during bacterial infections via its anti-inflammatory and antibacterial properties, PPARγ agonists have also been shown to dampen the host immune response and in some cases exacerbate infection by promoting leukocyte apoptosis and interfering with leukocyte migration and infiltration. In this review we discuss the role of PPARγ and its activation during bacterial infections, with focus on the potential of PPARγ agonists and perhaps antagonists as novel therapeutic modalities. We conclude that adjustment in the dosage and timing of PPARγ agonist administration, based on the competence of host antimicrobial defenses and the extent of inflammatory response and tissue injury, is critical for achieving the essential balance between pro- and anti-inflammatory effects on the immune system.
Collapse
|
12
|
Rajasekaran G, Kamalakannan R, Shin SY. Enhancement of the anti-inflammatory activity of temporin-1Tl-derived antimicrobial peptides by tryptophan, arginine and lysine substitutions. J Pept Sci 2015; 21:779-785. [DOI: doi 10.1002/psc.2807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Affiliation(s)
- Ganesan Rajasekaran
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
| | | | - Song Yub Shin
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
- Department of Cellular and Molecular Medicine, School of Medicine; Chosun University; Gwangju 501-759 Korea
| |
Collapse
|
13
|
Rajasekaran G, Kamalakannan R, Shin SY. Enhancement of the anti-inflammatory activity of temporin-1Tl-derived antimicrobial peptides by tryptophan, arginine and lysine substitutions. J Pept Sci 2015; 21:779-85. [DOI: 10.1002/psc.2807] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/10/2015] [Accepted: 07/10/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Ganesan Rajasekaran
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
| | | | - Song Yub Shin
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
- Department of Cellular and Molecular Medicine, School of Medicine; Chosun University; Gwangju 501-759 Korea
| |
Collapse
|
14
|
Dhama K, Saminathan M, Jacob SS, Singh M, Karthik K, . A, Tiwari R, Sunkara LT, Malik YS, Singh RK. Effect of Immunomodulation and Immunomodulatory Agents on Health with some Bioactive Principles, Modes of Action and Potent Biomedical Applications. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.253.290] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Romani L, Puccetti P. Controlling pathogenic inflammation to fungi. Expert Rev Anti Infect Ther 2014; 5:1007-17. [DOI: 10.1586/14787210.5.6.1007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
16
|
Cirioni O, Silvestri C, Pierpaoli E, Barucca A, Kamysz W, Ghiselli R, Scalise A, Brescini L, Castelli P, Orlando F, Kamysz E, Guerrieri M, Giacometti A, Provinciali M. IB-367 pre-treatment improves the in vivo efficacy of teicoplanin and daptomycin in an animal model of wounds infected with meticillin-resistant Staphylococcus aureus. J Med Microbiol 2013; 62:1552-1558. [DOI: 10.1099/jmm.0.057414-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antimicrobial peptides are known as immunomodulators and antibiotic enhancers. We report that administration of an antimicrobial peptide, IB-367, was efficacious in increasing the antimicrobial activity of daptomycin and teicoplanin in a mouse model of wound infection caused by meticillin-resistant Staphylococcus aureus (MRSA). Mice were assigned to seven groups: an IB-367 pre-treated group with no antibiotics given after challenge, two IB-367 pre-treated groups plus daptomycin or teicoplanin given after challenge, two groups treated with daptomycin or teicoplanin only after challenge, and two control groups without infection or that did not receive any treatment. The main outcome measures were quantitative bacterial culture and analysis of natural killer (NK) cytotoxicity and leukocyte phenotype. The wound, established through the panniculus carnosus muscle of mice, was infected by MRSA. Bacterial cultures of mice receiving antibiotics alone showed a −2 log decrease, whilst those for IB-367 plus daptomycin or teicoplanin showed a −4 log decrease. IB-367 plus daptomycin showed the highest efficacy. The higher antimicrobial effect exerted by IB-367 was associated with increased levels of NK cytotoxicity but not of NK cell number. IB-367 increased the number of both CD11b and Gr-1 cells 3 days after MRSA challenge, whereas both of these leukocyte populations were reduced at 10 days after challenge. Our data suggest that a combination of IB-367 with antibiotic exerts a therapeutic effect and may be useful for the management of staphylococcal wounds.
Collapse
Affiliation(s)
- Oscar Cirioni
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Carmela Silvestri
- Institute of Infectious Diseases and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Elisa Pierpaoli
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA – IRRCS, Ancona, Italy
| | - Alessandra Barucca
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA – IRRCS, Ancona, Italy
| | - Wojciech Kamysz
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - 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
| | - Pamela Castelli
- 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 – IRRCS, 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
| | - Mauro Provinciali
- Experimental Animal Models for Aging Unit, Scientific Technological Area, INRCA – IRRCS, Ancona, Italy
| |
Collapse
|
17
|
Hu L, Sun C, Wang S, Su F, Zhang S. Lipopolysaccharide neutralization by a novel peptide derived from phosvitin. Int J Biochem Cell Biol 2013; 45:2622-31. [PMID: 24028820 DOI: 10.1016/j.biocel.2013.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/27/2013] [Accepted: 09/02/2013] [Indexed: 11/25/2022]
Abstract
Lipopolysaccharide (LPS), also known as endotoxin, is the primary trigger of sepsis, which is associated with high mortality in patients. No therapeutic agents are currently efficacious enough to protect patients from sepsis characterized by LPS-mediated tissue damage and organ failure. Previously, a phosvitin-derived peptide, Pt5, which consists of the C-terminal 55 residues of zebrafish phosvitin, has been shown to function as an antibacterial agent. In this study, we have generated six mutants by site-directed mutagenesis based on the sequence of Pt5, and found that one of the six mutants, Pt5e, showed the strongest bactericidal activities against Escherichia coli and Staphylococcus aureus. We then demonstrated that Pt5e was able to bind to LPS and lipoteichoic acid (LTA). More importantly, we showed that Pt5e significantly inhibited LPS-induced tumor-necrosis factor (TNF)-α and interleukin (IL)-1β release from murine RAW264.7 cells and considerably reduced serum TNF-α and IL-1β levels in mice. Additionally, Pt5e protected the liver from damage by LPS, and remarkably promoted the survival rate of the endotoxemia mice. Furthermore, Pt5e displayed no cytotoxicity to murine RAW264.7 macrophages and no hemolytic activity toward human red blood cells. These data together indicate that Pt5e is an endotoxin-neutralizing agent with a therapeutic potential in clinical treatment of LPS-induced sepsis.
Collapse
Affiliation(s)
- Lili Hu
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | | | | | | | | |
Collapse
|
18
|
Ingle AM, Verma AK, Tiwari R, Karthik K, Chakraborty S, Deb R, Rajagunalan S, Rathore R, Dhama K. Immunomodulators in day to day life: a review. Pak J Biol Sci 2013; 16:826-843. [PMID: 24498836 DOI: 10.3923/pjbs.2013.826.843] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
There are ongoing trends of immunomodulation to combat a vast range of human and animal diseases including the incurable diseases like viral diseases, cancers, autoimmune diseases and inflammatory conditions. Animate as well as non-animate factors, surrounding us are interacting with our immune system. A balanced diet should contain all essential components from energy to vitamin and trace minerals. Each of these constituent has a very special effect on the immune system starting from their development to active role in immunity therefore, the outcome of their deficiency often ends in disease. Edible items which we consume like various vegetables, spices, herbs, fruits etc., are also equally responsible in manipulation of our system either in positive or negative way. Water has biggest share in our body and acts as the main medium to support the activities of the different system of body without exception of immune system. Proper environmental temperature is essential to maintain body's functions and experiments carried out regarding the effect of temperature suggest that extremes of the temperature are often cause immunosuppression directly by acting on the cells of immunity or indirectly through inducing stress and thereby increasing production of catecholamine which are potent anti-immune molecules. Various pathogenic as well as non-pathogenic bacteria cause immune suppression and immune potentiation, respectively. Proper exercise hold a prime position in the healthy life as it supports immunity and keeps disease away. The present review deals with all these immunomodulators having both positive and negative impact on the health status of an individual.
Collapse
Affiliation(s)
- Abhijeet M Ingle
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Izatnagar, Bareilly (UP)-243122, India
| | - Amit Kumar Verma
- Department of Veterinary Epidemiology and Preventive Medicine, Uttar Pradesh Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Viswavidyalaya Evum Go-Anusandhan Sansthan, Mathura-281001, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, Uttar Pradesh Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Viswavidyalaya Evum Go-Anusandhan Sansthan, Mathura-281001, India
| | - K Karthik
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Izatnagar, Bareilly (UP)-243122, India
| | - Sandip Chakraborty
- Deaprtment of Animal Resource Development, Pt. Nehru Complex, Agartala, Tripura-799001, India
| | - Rajib Deb
- Animal Genetics and Breeding, Project Directorate on Cattle, Indian Council of Agricultural Research, Grass Farm Road, Meerut, (UP)-250001, India
| | - S Rajagunalan
- Division of Veterinary Public Health, Indian Veterinary Research Institute, Izatnagar, Bareilly (UP)-243122, India
| | - Rajesh Rathore
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Izatnagar, Bareilly (UP)-243122, India
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute, Izatnagar, Bareilly (UP)-243122, India
| |
Collapse
|
19
|
Ding Y, Liu X, Bu L, Li H, Zhang S. Antimicrobial-immunomodulatory activities of zebrafish phosvitin-derived peptide Pt5. Peptides 2012; 37:309-13. [PMID: 22841856 DOI: 10.1016/j.peptides.2012.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 07/18/2012] [Accepted: 07/18/2012] [Indexed: 02/04/2023]
Abstract
A phosvitin (Pv)-derived peptide, Pt5, which consists of the C-terminal 55 residues of Pv in zebrafish, has been shown to function as an antimicrobial agent capable of killing microbes in vitro. However, its in vivo role in zebrafish remains unknown. In this study, we clearly demonstrated that Pt5 protected adult zebrafish from pathogenic Aeromonas hydrophila attack, capable of significantly enhancing the survival rate of zebrafish after the pathogenic challenge. Pt5 also caused a marked decrease in the numbers of A. hydrophila in the blood, spleen, kidney, liver and muscle, suggesting that Pt5 was able to block multiplication/dissemination of A. hydrophila in zebrafish. Additionally, Pt5 markedly suppressed the expression of the proinflammatory cytokine genes IL-1β, IL-6, TNF-α and IFN-γ in the spleen and head kidney of A. hydrophila-infected zebrafish, but it considerably enhanced the expressions of the antiinflammatory cytokine genes IL-10 and IL-4 in the same tissues. Taken together, these data indicate that Pt5 plays a dual role in zebrafish as an antimicrobial and immunomodulatory agent, capable of protecting zebrafish against pathogenic A. hydrophila through its antimicrobial activity as well as preventing zebrafish from the detrimental effects of an excessive inflammatory response via modulating immune functions.
Collapse
Affiliation(s)
- Yunchao Ding
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao 266003, China
| | | | | | | | | |
Collapse
|
20
|
Abstract
Despite appropriate antifungal treatment, the management of cryptococcal disease remains challenging, especially in immunocompromised patients, such as human immunodeficiency virus-infected individuals and solid organ transplant recipients. During the past two decades, our knowledge of host immune responses against Cryptococcus spp. has been greatly advanced, and the role of immunomodulation in augmenting the response to infection has been investigated. In particular, the role of 'protective' Th1 (tumour necrosis factor-α, interferon (IFN)-γ, interleukin (IL)-12, and IL-18) and Th17 (IL-23 and IL-17) and 'non-protective' Th2 (IL-4, IL-10, and IL-13) cytokines has been extensively studied in vitro and in animal models of cryptococcal infection. Immunomodulation with monoclonal antibodies against the capsular polysaccharide glucuronoxylomannan, glucosylceramides, melanin and β-glucan and, lately, with radioimmunotherapy has also yielded promising results in animal models. As a balance between sufficiently protective Th1 responses and excessive inflammation is important for optimal outcome, the effect of immunotherapy may range from beneficial to deleterious, depending on factors related to the host, the infecting organism, and the immunomodulatory regimen. Clinical evidence supporting immunomodulation in patients with cryptococcal infection remains too limited to allow firm recommendations. Limited human data suggest a role for IFN-γ. Identification of surrogate markers characterizing patients' immunological status could possibly suggest candidate patients for immunotherapy and the type of immunomodulation to be administered.
Collapse
Affiliation(s)
- C Antachopoulos
- 3rd Department of Paediatrics, Hippokration Hospital, Aristotle University, Thessaloniki, Greece
| | | |
Collapse
|
21
|
|
22
|
Smit E, Oberholzer HM, Pretorius E. A review of immunomodulators with reference to Canova. HOMEOPATHY 2009; 98:169-76. [PMID: 19647212 DOI: 10.1016/j.homp.2009.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 04/30/2009] [Accepted: 05/05/2009] [Indexed: 11/29/2022]
Abstract
Immunomodulators are substances which modify the immunity of an individual to favour a particular immunological response. The immune response and the function of the immune response regulation process are described, with special reference to cancer and autoimmune disease. Homeopathy and its role in immune regulation are discussed with special reference to Canova. Canova is a homeopathic product produced, according to the Hahnemannian homeopathic method, in Brazil. Its role in cancer, bone marrow and haematopoiesis as well as macrophage and monocyte activation is reviewed. Canova seems to stabilize platelet morphology in human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS). The data suggest that the future of immunomodulators and homeopathic products which appear to have an effect on the immune response requires a better understanding of the relative need for immune activation versus immune modulation. Homeopathic products specifically need more attention.
Collapse
Affiliation(s)
- E Smit
- Department of Anatomy, University of Pretoria, Pretoria, Gauteng 0001, South Africa
| | | | | |
Collapse
|
23
|
Abstract
Protective immunity against fungal pathogens is achieved by the integration of two distinct arms of the immune system, the innate and adaptive responses. Innate and adaptive immune responses are intimately linked and controlled by sets of molecules and receptors that act to generate the most effective form of immunity for protection against fungal pathogens. The decision of how to respond will still be primarily determined by interactions between pathogens and cells of the innate immune system, but the actions of T cells will feed back into this dynamic equilibrium to regulate the balance between tolerogenic and inflammatory responses. In the last two decades, the immunopathogenesis of fungal infections and fungal diseases was explained primarily in terms of Th1/Th2 balance. Although Th1 responses driven by the IL-12/IFN-gamma axis are central to protection against fungi, other cytokines and T cell-dependent pathways have come of age. The newly described Th17 developmental pathway may play an inflammatory role previously attributed to uncontrolled Th1 responses and serves to accommodate the seemingly paradoxical association of chronic inflammatory responses with fungal persistence in the face of an ongoing inflammation. Regulatory T cells in their capacity to inhibit aspects of innate and adaptive antifungal immunity have become an integral component of immune resistance to fungi, and provide the host with immune defense mechanisms adequate for protection, without necessarily eliminating fungal pathogens which would impair immune memory--or causing an unacceptable level of tissue damage. The enzyme indoleamine 2,3-dioxygenase and tryptophan metabolites contribute to immune homeostasis by inducing Tregs and taming overzealous or heightened inflammatory responses.
Collapse
Affiliation(s)
- Luigina Romani
- Microbiology Section, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy.
| |
Collapse
|
24
|
Cegelski L, Marshall GR, Eldridge GR, Hultgren SJ. The biology and future prospects of antivirulence therapies. Nat Rev Microbiol 2008; 6:17-27. [PMID: 18079741 DOI: 10.1038/nrmicro1818] [Citation(s) in RCA: 579] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The emergence and increasing prevalence of bacterial strains that are resistant to available antibiotics demand the discovery of new therapeutic approaches. Targeting bacterial virulence is an alternative approach to antimicrobial therapy that offers promising opportunities to inhibit pathogenesis and its consequences without placing immediate life-or-death pressure on the target bacterium. Certain virulence factors have been shown to be potential targets for drug design and therapeutic intervention, whereas new insights are crucial for exploiting others. Targeting virulence represents a new paradigm to empower the clinician to prevent and treat infectious diseases.
Collapse
Affiliation(s)
- Lynette Cegelski
- Department of Molecular Microbiology, Washington University, Saint Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
25
|
Pirofski LA, Casadevall A. The damage-response framework of microbial pathogenesis and infectious diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 635:135-46. [PMID: 18841709 PMCID: PMC7123708 DOI: 10.1007/978-0-387-09550-9_11] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Historical and most currently held views of microbial pathogenesis and virulence are plagued by confusing and imprecise terminology and definitions that require revision and exceptions to accommodate new basic science and clinical information about microbes and infectious diseases. These views are also inherently unable to account for the ability of some microbes to cause disease in certain, but not other hosts, because they are grounded in singular, either microbe-or host-centric views. The damage-response framework is an integrated theory of microbial pathogenesis that puts forth the view that microbial pathogenesis reflects the outcome of an interaction between a host and a microbe, with each entity contributing to the nature of the outcome, which in turn depends on the amount of host damage that results from the host-microbe interaction. This view is able to accommodate new information and explain why infection with the same microbe can have different outcomes in different hosts. This chapter describes the origins and conceptual underpinnings of and the outcomes of infection put forth in, the damage-response framework.
Collapse
Affiliation(s)
- Liise-anne Pirofski
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
26
|
Fabrizio K, Groner A, Boes M, Pirofski LA. A human monoclonal immunoglobulin M reduces bacteremia and inflammation in a mouse model of systemic pneumococcal infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:382-90. [PMID: 17301214 PMCID: PMC1865609 DOI: 10.1128/cvi.00374-06] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Antibody-based approaches to pneumococcal disease may hold promise for immunocompromised patients in whom vaccines are less immunogenic and/or in the context of antimicrobial resistance. Antibody-mediated protection against experimental pneumococcal pneumonia has been shown to depend on immunoregulation, but the relationship between antibody and protection against pneumococcal sepsis and immunoregulation has not been examined. Similarly, the requirement for B and T cells for antibody efficacy is not known. In this study, we determined the efficacy of the human pneumococcal capsular polysaccharide serotype 3-specific antibody, A7 (immunoglobulin M [IgM]), in secretory IgM (sIgM)(-/-), CD4(-/-), CD8(-/-), muMT(-/-), and SCID mice and investigated its effect on cytokine and chemokine expression in sera and spleens from mice with intact cellular immunity. A7 is known to be protective against systemic infection with serotype 3 and to require complement for efficacy. Compared to that of an isotype control antibody, A7 administration prolonged the survival of mice of each immunodeficient strain and was associated with a significant reduction in CFU in blood, lung, and spleen samples and a significantly reduced level of keratinocyte-derived chemokine (KC), interleukin-6 (IL-6), and macrophage inflammatory protein-2 (MIP-2) expression in normal and sIgM(-/-) mice. Studies with mice treated with penicillin revealed similar reductions in CFU and similar levels of IL-6, KC, or MIP-2 expression in A7- and penicillin-treated mice. These findings demonstrate that natural IgM and B and T cells are dispensable for A7-mediated protection against experimental pneumococcal sepsis and suggest that the efficacy of antibody-mediated protection depends on immunomodulation. Taken together, our data extend the association between antibody-mediated protection and immunomodulation to protection against systemic pneumococcal infection and to a clinically important serotype often responsible for pneumococcal sepsis.
Collapse
Affiliation(s)
- Kevin Fabrizio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
27
|
Biziulevicius GA, Kazlauskaite J. Pushing ‘bad bugs’ into committing suicide: Activation of microbial autolysis within the gut using food-grade substances as a prospective method for treatment of intestinal infections and/or immunity enhancement. Med Hypotheses 2007; 69:1161-2. [PMID: 17418499 DOI: 10.1016/j.mehy.2007.01.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Accepted: 01/27/2007] [Indexed: 11/25/2022]
|
28
|
|