1
|
Mejía SP, Marques RDC, Landfester K, Orozco J, Mailänder V. Effect of Protein Corona on the Specificity and Efficacy of Nanobioconjugates to Treat Intracellular Infections. Macromol Biosci 2024; 24:e2300197. [PMID: 37639236 DOI: 10.1002/mabi.202300197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/20/2023] [Indexed: 08/29/2023]
Abstract
Encapsulating drugs into functionalized nanoparticles (NPs) is an alternative to reach the specific therapeutic target with lower doses. However, when the NPs are in contact with physiological media, proteins adsorb on their surfaces, forming a protein corona (PC) biomolecular layer, acquiring a distinct biological identity that alters their interactions with cells. Itraconazole (ITZ), an antifungal agent, is encapsulated into PEGylated and/or functionalized NPs with high specificity for macrophages. It is evaluated how the PC impacts their cell uptake and antifungal effect. The minimum inhibitory concentration and colony-forming unit assays demonstrate that encapsulated ITZ into poly(ethylene glycol) (PEG) NPs improves the antifungal effect compared with NPs lacking PEGylation. The improvement can be related to the synergistic effect of the encapsulated ITZ and NPs composition and the reduction of PC formation in PEG NPs. Functionalized NPs with anti-F4/80 and anti-MARCO antibodies, or mannose without PEG and treated with PC, show an improved uptake but, in the presence of PEG, significantly reduce the endocytosis, dominating the stealth effect from PEG. Therefore, the PC plays a crucial role in the nanosystem uptake and antifungal effects, which suggests the need for in vivo model studies to evaluate the effect of PC in the specificity and biodistribution.
Collapse
Affiliation(s)
- Susana P Mejía
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 No 52-20, Medellin, 050010, Colombia
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | | | | | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 No 52-20, Medellin, 050010, Colombia
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeck str. 1, 55131, Mainz, Germany
| |
Collapse
|
2
|
Tian N, Duan H, Cao T, Dai G, Sheng G, Chu H, Sun Z. Macrophage-targeted nanoparticles mediate synergistic photodynamic therapy and immunotherapy of tuberculosis. RSC Adv 2023; 13:1727-1737. [PMID: 36712647 PMCID: PMC9832440 DOI: 10.1039/d2ra06334d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) that poses a serious global public health threat. Due to the high incidence of adverse reactions associated with conventional treatment regimens, there is an urgent need for better alternative therapies. CpG oligodeoxynucleotides (CpG ODNs) are synthetic oligodeoxyribonucleotide sequences. They can induce a Th1-type immune response by stimulating Toll-like receptors (TLRs) in mammalian immune cells, thus killing Mtb. However, due to the negative charge and easy degradation of CpG ODNs, it is necessary to deliver them into cells using nanomaterials. PCN-224 (hereinafter referred to as PCN), as a metal-organic framework based on zirconium ions and porphyrin ligands, not only has the advantage of high drug loading capacity, but also the porphyrin molecule in it is a type of photosensitizer, which allows these nanocomposites to play a role in photodynamic therapy (PDT) while delivering CpG ODNs. In addition, since Mtb mainly exists in macrophages, targeting anti-TB agents to macrophages is helpful to improve the anti-TB effect. Phosphatidylserine (PS) is a biological membrane phospholipid that is normally found on the inner side of cell membranes in, for example, plant and mammalian cells. When apoptosis occurs, PS can flip from the inner side of the cell membrane to the surface of the cell membrane, displaying a specific "eat-me" signal that can be recognized by specific receptors on macrophages. Therefore, we can use this macrophage-targeting property of PS to construct bio-inspired targeted drug delivery systems. In this study, we constructed PCN-CpG@PS nanocomposites. PCN-CpG@PS, combining PDT and immunotherapy, is designed to target macrophages at the site of a lesion and kill latent Mtb. We physically characterized the nanocomposites and validated their bactericidal ability in vitro and their ability to stimulate the immune system in vivo. The results demonstrated that the targeted nanocomposites have certain in vitro antituberculosis efficacy with good safety.
Collapse
Affiliation(s)
- Na Tian
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Huijuan Duan
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Tingming Cao
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Guangming Dai
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Gang Sheng
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Hongqian Chu
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| | - Zhaogang Sun
- Beijing Chest Hospital, Capital Medical University Beijing 101149 China
- Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Institute Beijing 101149 China
| |
Collapse
|
3
|
Pereira de Sa N, Del Poeta M. Sterylglucosides in Fungi. J Fungi (Basel) 2022; 8:1130. [PMID: 36354897 PMCID: PMC9698648 DOI: 10.3390/jof8111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Sterylglucosides (SGs) are sterol conjugates widely distributed in nature. Although their universal presence in all living organisms suggests the importance of this kind of glycolipids, they are yet poorly understood. The glycosylation of sterols confers a more hydrophilic character, modifying biophysical properties of cell membranes and altering immunogenicity of the cells. In fungi, SGs regulate different cell pathways to help overcome oxygen and pH challenges, as well as help to accomplish cell recycling and other membrane functions. At the same time, the level of these lipids is highly controlled, especially in wild-type fungi. In addition, modulating SGs metabolism is becoming a novel tool for vaccine and antifungal development. In the present review, we bring together multiple observations to emphasize the underestimated importance of SGs for fungal cell functions.
Collapse
Affiliation(s)
- Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
- Institute of Chemical Biology and Drug Discovery (ICB&DD), Stony Brook, NY 11794, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Veterans Administration Medical Center, Northport, NY 11768, USA
| |
Collapse
|
4
|
Antifungal Encapsulated into Ligand-Functionalized Nanoparticles with High Specificity for Macrophages. Pharmaceutics 2022; 14:pharmaceutics14091932. [PMID: 36145686 PMCID: PMC9501281 DOI: 10.3390/pharmaceutics14091932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/28/2022] Open
Abstract
Infectious diseases caused by intracellular microorganisms such as Histoplasma capsulatum represent a significant challenge worldwide. Drug encapsulation into functionalized nanoparticles (NPs) is a valuable alternative to improving drug solubility and bioavailability, preventing undesirable interactions and drug degradation, and reaching the specific therapeutic target with lower doses. This work reports on Itraconazole (ITZ) encapsulated into core-shell-like polymeric NPs and functionalized with anti-F4/80 antibodies for their targeted and controlled release into macrophages. Uptake assay on co-culture showed significant differences between the uptake of functionalized and bare NPs, higher with functionalized NPs. In vitro assays showed that F4/80-NPs with 0.007 µg/mL of encapsulated ITZ eliminated the H. capsulatum fungus in co-culture with macrophages effectively compared to the bare NPs, without any cytotoxic effect on macrophages after 24 h interaction. Furthermore, encapsulated ITZ modulated the gene expression of anti and pro-inflammatory cytokines (IL-1, INF-Y, IL-6 and IL-10) on macrophages. Additionally, the anti-F4/80 antibody-coating enhanced natural and adequate antifungal response in the cells, exerting a synergistic effect that prevented the growth of the fungus at the intracellular level. Functionalized NPs can potentially improve macrophage-targeted therapy, increasing NPs endocytosis and intracellular drug concentration.
Collapse
|
5
|
Nanomedicine Reformulation of Chloroquine and Hydroxychloroquine. Molecules 2020; 26:molecules26010175. [PMID: 33396545 PMCID: PMC7794963 DOI: 10.3390/molecules26010175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/18/2022] Open
Abstract
The chloroquine family of antimalarials has a long history of use, spanning many decades. Despite this extensive clinical experience, novel applications, including use in autoimmune disorders, infectious disease, and cancer, have only recently been identified. While short term use of chloroquine or hydroxychloroquine is safe at traditional therapeutic doses in patients without predisposing conditions, administration of higher doses and for longer durations are associated with toxicity, including retinotoxicity. Additional liabilities of these medications include pharmacokinetic profiles that require extended dosing to achieve therapeutic tissue concentrations. To improve chloroquine therapy, researchers have turned toward nanomedicine reformulation of chloroquine and hydroxychloroquine to increase exposure of target tissues relative to off-target tissues, thereby improving the therapeutic index. This review highlights these reformulation efforts to date, identifying issues in experimental designs leading to ambiguity regarding the nanoformulation improvements and lack of thorough pharmacokinetics and safety evaluation. Gaps in our current understanding of these formulations, as well as recommendations for future formulation efforts, are presented.
Collapse
|
6
|
Rossato L, Camargo Dos Santos M, Vitale RG, de Hoog S, Ishida K. Alternative treatment of fungal infections: Synergy with non-antifungal agents. Mycoses 2020; 64:232-244. [PMID: 33098146 DOI: 10.1111/myc.13203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Fungal infections are responsible for high mortality rates in immunocompromised and high-risk surgical patients. Therapy failures during the last decades due to increasing multidrug resistance demand innovative strategies for novel and effective antifungal drugs. Synergistic combinations of antifungals with non-antifungal agents highlight a pragmatic strategy to reduce the development of drug resistance and potentially repurpose known compounds with other functions to bypass costly and time-consuming novel drug development.
Collapse
Affiliation(s)
- Luana Rossato
- Faculdade de Ciências da Saúde, Federal University of Grande Dourados, Mato Grosso do Sul, Brazil
| | | | - Roxana G Vitale
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) and Hospital JM Ramos Mejía, Ciudad Autónoma de Buenos Aires, Argentina
| | - Sybren de Hoog
- Center of Expertise in Mycology of Radboud University Medical Center, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Normile TG, Bryan AM, Del Poeta M. Animal Models of Cryptococcus neoformans in Identifying Immune Parameters Associated With Primary Infection and Reactivation of Latent Infection. Front Immunol 2020; 11:581750. [PMID: 33042164 PMCID: PMC7522366 DOI: 10.3389/fimmu.2020.581750] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Cryptococcus species are environmental fungal pathogens and the causative agents of cryptococcosis. Infection occurs upon inhalation of infectious particles, which proliferate in the lung causing a primary infection. From this primary lung infection, fungal cells can eventually disseminate to other organs, particularly the brain, causing lethal meningoencephalitis. However, in most cases, the primary infection resolves with the formation of a lung granuloma. Upon severe immunodeficiency, dormant cryptococcal cells will start proliferating in the lung granuloma and eventually will disseminate to the brain. Many investigators have sought to study the protective host immune response to this pathogen in search of host parameters that keep the proliferation of cryptococcal cells under control. The majority of the work assimilates research carried out using the primary infection animal model, mainly because a reactivation model has been available only very recently. This review will focus on anti-cryptococcal immunity in both the primary and reactivation models. An understanding of the differences in host immunity between the primary and reactivation models will help to define the key host parameters that control the infections and are important for the research and development of new therapeutic and vaccine strategies against cryptococcosis.
Collapse
Affiliation(s)
- Tyler G Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Arielle M Bryan
- Ingenious Targeting Laboratory Incorporated, Ronkonkoma, NY, United States
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States.,Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY, United States.,Veterans Administration Medical Center, Northport, NY, United States
| |
Collapse
|
8
|
Khan MA, Khan A, Khan SH, Azam M, Khan MMU, Khalilullah H, Younus H. Coadministration of liposomal methylglyoxal increases the activity of amphotericin B against Candida albicans in leukopoenic mice. J Drug Target 2020; 29:78-87. [PMID: 32723117 DOI: 10.1080/1061186x.2020.1803333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this study, we investigated the therapeutic efficacy of a combination of liposomal amphotericin B (Lip-Amp B) and Methylglyoxal (Lip-MG) against Candida albicans in the leukopoenic mice. The antifungal efficacy of Lip-Amp B or Lip-MG or a combination of Lip-Amp B and Lip-MG was evaluated by the analysis of the survival rate and the fungal load in the treated mice. The immune-stimulatory effect of Lip-MG on macrophages was evaluated by analysing the secretion of proinflammatory cytokines. C. albicans infected mice treated at the doses of 1 and 2 mg/kg of Lip-Amp B showed 20% and 50% survival rates, respectively. Whereas the mice treated with free Amp B at the same doses died within 40 days of treatment. Interestingly, C. albicans infected mice treated with a combination of Lip-Amp B and Lip-MG had 70% survival rate on day 40 postinfection. Moreover, treatment of macrophages with Lip-MG increased their fungicidal activity and the secretion of proinflammatory cytokines, including TNF-α and IL-1β. These findings suggested that co-treatment with Lip-Amp B and Lip-MG had a synergistic effect and could be effective against C. albicans in immunocompromised subjects.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Mohd Azam
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Mohd Masih Uzzaman Khan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Saudi Arabia
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Saudi Arabia
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
9
|
Khan SH, Younus H, Allemailem KS, Almatroudi A, Alrumaihi F, Alruwetei AM, Alsahli MA, Khan A, Khan MA. Potential of Methylglyoxal-Conjugated Chitosan Nanoparticles in Treatment of Fluconazole-Resistant Candida albicans Infection in a Murine Model. Int J Nanomedicine 2020; 15:3681-3693. [PMID: 32547022 PMCID: PMC7261666 DOI: 10.2147/ijn.s249625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fungal infections are becoming more prevalent and threatening because of the continuous emergence of azole-resistant fungal infections. The present study was aimed to assess the activity of free Methylglyoxal (MG) or MG-conjugated chitosan nanoparticles (MGCN) against fluconazole-resistant Candida albicans. MATERIALS AND METHODS A novel formulation of MGCN was prepared and characterized to determine their size, shape and polydispersity index. Moreover, the efficacy of fluconazole or MG or MGCN was determined against intracellular C. albicans in macrophages and the systematic candidiasis in a murine model. The safety of MG or MGCN was tested in mice by analyzing the levels of hepatic and renal toxicity parameters. RESULTS Candida albicans did not respond to fluconazole, even at the highest dose of 20 mg/kg, whereas MG and MGCN effectively eliminated C. albicans from the macrophages and infected mice. Mice in the group treated with MGCN at a dose of 10 mg/kg exhibited a 90% survival rate and showed the lowest fungal load in the kidney, whereas the mice treated with free MG at the same dose exhibited 50% survival rate. Moreover, the administration of MG or MGCN did not induce any liver and kidney toxicity in the treated mice. CONCLUSION The findings of the present work suggest that MGCN may be proved a promising therapeutic formulation to treat azole-resistant C. albicans infections.
Collapse
Affiliation(s)
- Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh202002, India
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh202002, India
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Abdulmohsen M Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, 51452, Saudi Arabia
| |
Collapse
|
10
|
Nicola AM, Albuquerque P, Paes HC, Fernandes L, Costa FF, Kioshima ES, Abadio AKR, Bocca AL, Felipe MS. Antifungal drugs: New insights in research & development. Pharmacol Ther 2018; 195:21-38. [PMID: 30347212 DOI: 10.1016/j.pharmthera.2018.10.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The need for better antifungal therapy is commonly accepted in view of the high mortality rates associated with systemic infections, the low number of available antifungal classes, their associated toxicity and the increasing number of infections caused by strains with natural or acquired resistance. The urgency to expand the range of therapeutic options for the treatment of fungal infections has led researchers in recent decades to seek alternative antifungal targets when compared to the conventional ones currently used. Although new potential targets are reported, translating the discoveries from bench to bedside is a long process and most of these drugs fail to reach the patients. In this review, we discuss the development of antifungal drugs focusing on the approach of drug repurposing and the search for novel drugs for classical targets, the most recently described gene targets for drug development, the possibilities of immunotherapy using antibodies, cytokines, therapeutic vaccines and antimicrobial peptides.
Collapse
Affiliation(s)
| | - Patrícia Albuquerque
- Faculty of Ceilândia, University of Brasília, Brazil; Graduate Programme in Microbial Biology, University of Brasília, Brazil
| | - Hugo Costa Paes
- Division of Clinical Medicine, University of Brasília Medical School, Brazil
| | - Larissa Fernandes
- Faculty of Ceilândia, University of Brasília, Brazil; Graduate Programme in Microbial Biology, University of Brasília, Brazil
| | - Fabricio F Costa
- Graduate Programme in Genomic Science and Biotechnology, Catholic University of Brasília, Brazil; MATTER, Chicago, IL, USA; Cancer Biology and Epigenomics Program, Ann & Robert Lurie Children's Hospital of Chicago Research Center, Northwestern University's Feinberg School of Medicine, Chicago, Illinois, USA
| | - Erika Seki Kioshima
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Paraná, Brazil
| | - Ana Karina Rodrigues Abadio
- School for Applied Social and Agricultural Sciences, State University of Mato Grosso, Nova Mutum Campus, Mato Grosso, Brazil
| | | | - Maria Sueli Felipe
- Graduate Programme in Genomic Science and Biotechnology, Catholic University of Brasília, Brazil; Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brazil.
| |
Collapse
|
11
|
Folkerts H, Hilgendorf S, Vellenga E, Bremer E, Wiersma VR. The multifaceted role of autophagy in cancer and the microenvironment. Med Res Rev 2018; 39:517-560. [PMID: 30302772 PMCID: PMC6585651 DOI: 10.1002/med.21531] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is a crucial recycling process that is increasingly being recognized as an important factor in cancer initiation, cancer (stem) cell maintenance as well as the development of resistance to cancer therapy in both solid and hematological malignancies. Furthermore, it is being recognized that autophagy also plays a crucial and sometimes opposing role in the complex cancer microenvironment. For instance, autophagy in stromal cells such as fibroblasts contributes to tumorigenesis by generating and supplying nutrients to cancerous cells. Reversely, autophagy in immune cells appears to contribute to tumor‐localized immune responses and among others regulates antigen presentation to and by immune cells. Autophagy also directly regulates T and natural killer cell activity and is required for mounting T‐cell memory responses. Thus, within the tumor microenvironment autophagy has a multifaceted role that, depending on the context, may help drive tumorigenesis or may help to support anticancer immune responses. This multifaceted role should be taken into account when designing autophagy‐based cancer therapeutics. In this review, we provide an overview of the diverse facets of autophagy in cancer cells and nonmalignant cells in the cancer microenvironment. Second, we will attempt to integrate and provide a unified view of how these various aspects can be therapeutically exploited for cancer therapy.
Collapse
Affiliation(s)
- Hendrik Folkerts
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Susan Hilgendorf
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edwin Bremer
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Valerie R Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Biological macromolecules based targeted nanodrug delivery systems for the treatment of intracellular infections. Int J Biol Macromol 2018; 110:2-6. [PMID: 29355638 DOI: 10.1016/j.ijbiomac.2018.01.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 01/18/2023]
Abstract
Intracellular infections are tricky to treat, the reason being the poor penetration of antibiotics/antimycotics into the microbial niche (host cell). Macrophages are primary targets of facultative and obligate intracellular bacteria/fungi to be abused as host cells. The need for drugs with better intracellular penetration led to the development of endocytosable drug carriers, which can cross the cell membrane of the host cells (macrophages) by imitating the entry path of the pathogens. Therefore, the drugs can be targeted to macrophages ensuring enhanced therapeutic effect. This review discusses the exploitation of various nanocarriers for targeted delivery of drugs to the macrophages in the last two decades.
Collapse
|
13
|
Immune-Stimulatory and Therapeutic Activity of Tinospora cordifolia: Double-Edged Sword against Salmonellosis. J Immunol Res 2017; 2017:1787803. [PMID: 29318160 PMCID: PMC5727750 DOI: 10.1155/2017/1787803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/17/2017] [Indexed: 11/17/2022] Open
Abstract
The present study was aimed at determining the activity of aqueous and methanolic extracts of Tinospora cordifolia (AETC and METC) against Salmonella typhimurium. In vitro anti-Salmonella activity of T. cordifolia was determined through the broth dilution and agar well diffusion assays. The immune-stimulating potential of AETC or METC was determined by measuring the cytokine levels in the culture supernatants of treated murine J774 macrophages. Antibacterial activity of AETC or METC was determined by treating S. typhimurium-infected macrophages and BALB/C mice. The toxicity of AETC or METC was determined by measuring the levels of liver inflammation markers aspartate transaminase (AST) and alanine transaminase (ALT) and antioxidant enzymes. Macrophages treated with AETC or METC secreted greater levels of IFN-γ, TNF-α, and IL-1β. METC showed greater activity against S. typhimurium infection in macrophages and mice as well. Treatment with METC resulted in increased survival and reduced bacterial load in S. typhimurium-infected mice. Moreover, METC or AETC treatment reduced the liver inflammation and rescued the levels of antioxidant enzymes in S. typhimurium-infected mice. The results of the present study suggest that the use of T. cordifolia may act as a double-edged sword in combating salmonellosis.
Collapse
|
14
|
Wang J, Kang YX, Pan W, Lei W, Feng B, Wang XJ. Enhancement of Anti-Inflammatory Activity of Curcumin Using Phosphatidylserine-Containing Nanoparticles in Cultured Macrophages. Int J Mol Sci 2016; 17:ijms17060969. [PMID: 27331813 PMCID: PMC4926501 DOI: 10.3390/ijms17060969] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/03/2016] [Accepted: 06/09/2016] [Indexed: 12/16/2022] Open
Abstract
Macrophages are one kind of innate immune cells, and produce a variety of inflammatory cytokines in response to various stimuli, such as oxidized low density lipoprotein found in the pathogenesis of atherosclerosis. In this study, the effect of phosphatidylserine on anti-inflammatory activity of curcumin-loaded nanostructured lipid carriers was investigated using macrophage cultures. Different amounts of phosphatidylserine were used in the preparation of curcumin nanoparticles, their physicochemical properties and biocompatibilities were then compared. Cellular uptake of the nanoparticles was investigated using a confocal laser scanning microscope and flow cytometry analysis in order to determine the optimal phosphatidylserine concentration. In vitro anti-inflammatory activities were evaluated in macrophages to test whether curcumin and phosphatidylserine have interactive effects on macrophage lipid uptake behavior and anti-inflammatory responses. Here, we showed that macrophage uptake of phosphatidylserine-containing nanostructured lipid carriers increased with increasing amount of phosphatidylserine in the range of 0%–8%, and decreased when the phosphatidylserine molar ratio reached over 12%. curcumin-loaded nanostructured lipid carriers significantly inhibited lipid accumulation and pro-inflammatory factor production in cultured macrophages, and evidently promoted release of anti-inflammatory cytokines, when compared with curcumin or phosphatidylserine alone. These results suggest that the delivery system using PS-based nanoparticles has great potential for efficient delivery of drugs such as curcumin, specifically targeting macrophages and modulation of their anti-inflammatory functions.
Collapse
Affiliation(s)
- Ji Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| | - Yu-Xia Kang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| | - Wen Pan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| | - Wan Lei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| | - Bin Feng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| | - Xiao-Juan Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
15
|
Coelho C, Casadevall A. Cryptococcal therapies and drug targets: the old, the new and the promising. Cell Microbiol 2016; 18:792-9. [PMID: 26990050 DOI: 10.1111/cmi.12590] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/28/2016] [Accepted: 03/08/2016] [Indexed: 01/09/2023]
Abstract
Half a century after the introduction of Amphotericin B the management of cryptococcosis remains unsatisfactory. The disease, caused primarily by the two fungal species Cryptococcus neoformans and Cryptococcus gattii, remains responsible for considerable morbidity and mortality despite standard medical care. Current therapeutic options are limited to Amphotericin B, azoles and 5-flucytosine. However, this organism has numerous well-characterized virulence mechanisms that are amenable to pharmacological interference and are thus potential therapeutic targets. Here, we discuss existing approved antifungal drugs, resistance mechanisms to these drugs and non-standard antifungal drugs that have potential in treatment of cryptococcosis, including immunomodulatory strategies that synergize with antifungal drugs, such as cytokine administration or monoclonal antibodies. Finally, we summarize attempts to target well-described virulence factors of Cryptococcus, the capsule or fungal melanin. This review emphasizes the pressing need for new therapeutic alternatives for cryptococcosis.
Collapse
Affiliation(s)
- Carolina Coelho
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
16
|
Tong HI, Kang W, Shi Y, Zhou G, Lu Y. Physiological function and inflamed-brain migration of mouse monocyte-derived macrophages following cellular uptake of superparamagnetic iron oxide nanoparticles-Implication of macrophage-based drug delivery into the central nervous system. Int J Pharm 2016; 505:271-82. [PMID: 27001531 DOI: 10.1016/j.ijpharm.2016.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/05/2016] [Accepted: 03/18/2016] [Indexed: 02/05/2023]
Abstract
This study was designed to use superparamagnetic iron oxide nanoparticles (SPIONs) as evaluating tools to study monocyte-derived macrophages (MDM)-mediated delivery of small molecular agents into the diseased brains. MDM were tested with different-configured SPIONs at selected concentrations for their impacts on carrier cells' physiological and migratory properties, which were found to depend largely on particle size, coating, and treatment concentrations. SHP30, a SPION of 30-nm core size with oleic acids plus amphiphilic polymer coating, was identified to have high cellular uptake efficiency and cause little cytotoxic effects on MDM. At lower incubation dose (25μg/mL), few alteration was observed in carrier cells' physiological and in vivo migratory functions, as tested in a lipopolysaccharide-induced acute neuroinflammation mouse model. Nevertheless, significant increase in monocyte-to-macrophage differentiation, and decrease in in vivo carrier MDM inflamed-brain homing ability were found in groups treated with a higher dose of SHP30at 100μg/mL. Overall, our results have identified MDM treatment at 25μg/mL SHP30 resulted in little functional changes, provided valuable parameters for using SPIONs as evaluating tools to study MDM-mediated therapeutics carriage and delivery, and supported the concepts of using monocytes-macrophages as cellular vehicles to transport small molecular agents to the brain.
Collapse
Affiliation(s)
- Hsin-I Tong
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA; Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Wen Kang
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Yingli Shi
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Guangzhou Zhou
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Yuanan Lu
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| |
Collapse
|
17
|
Cavalcanti I, Pontes-Neto J, Kocerginsky P, Bezerra-Neto A, Lima J, Lira-Nogueira M, Maciel M, Neves R, Pimentel M, Santos-Magalhães N. Antimicrobial activity of β-lapachone encapsulated into liposomes against meticillin-resistant Staphylococcus aureus and Cryptococcus neoformans clinical strains. J Glob Antimicrob Resist 2015; 3:103-108. [DOI: 10.1016/j.jgar.2015.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 12/11/2022] Open
|
18
|
Srivastava V, Lee H. Chloroquine-based hybrid molecules as promising novel chemotherapeutic agents. Eur J Pharmacol 2015; 762:472-86. [PMID: 25959387 DOI: 10.1016/j.ejphar.2015.04.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/15/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
Chloroquine (CQ) has a broad spectrum of pharmacological activities including anticancer and anti-inflammatory, in addition to its well-known antimalarial activity. This very useful property of CQ may be rendered through a variety of different molecular and cellular mechanisms, including the induction of apoptosis, necrosis and lysosomal dysfunction. CQ alone may not be as effective as many well-known anticancer drugs; however, it often shows synergisticts when combined with other anticancer agents, without causing substantial ill-effects. To increase its pharmacological activity, scientists synthesized many different chloroquine derivatives by a repositioning approach, some of which show higher activities than the parental CQ. To further improve anticancer activity, medicinal chemists have recently been focusing on generating CQ hybrid molecules by joining, directly or through a linker, 4-aminoquinoline and other pharmacologically active phamarcophore(s). Indeed, some CQ hybrid molecules substantially improved anticancer activity while maintaining desirable CQ property, providing an excellent opportunity of developing effective and safe novel anticancer agents. Since the approach of developing CQ hybrid molecules has advanced much more in the antimalarial drug research, it can provide an excellent template for anticancer drug development. This review provides an overview of CQ-based hybrid molecules by focusing on: (1) the potential advantage of the hybrid approach in developing effective and safe anticancer agents; (2) what we can learn from the CQ hybrid approach used in the development of effective antimalarial agents; and (3) CQ hybrid molecules as potential anticancer agents in different categories classified based on their chemical compositions.
Collapse
Affiliation(s)
- Vandana Srivastava
- Advanced Medical Research Institute of Canada, Health Sciences North, 41 Ramsey Lake Road, Sudbury, Ontario, Canada P3E 5J1; Division of Medical Sciences, Northern Ontario School of Medicine, 935 Ramsey Lake Road, Sudbury, Ontario, Canada P3E 2C6
| | - Hoyun Lee
- Advanced Medical Research Institute of Canada, Health Sciences North, 41 Ramsey Lake Road, Sudbury, Ontario, Canada P3E 5J1; Division of Medical Sciences, Northern Ontario School of Medicine, 935 Ramsey Lake Road, Sudbury, Ontario, Canada P3E 2C6.
| |
Collapse
|
19
|
Dalhoff A. Antiviral, antifungal, and antiparasitic activities of fluoroquinolones optimized for treatment of bacterial infections: a puzzling paradox or a logical consequence of their mode of action? Eur J Clin Microbiol Infect Dis 2015; 34:661-8. [PMID: 25515946 PMCID: PMC7087824 DOI: 10.1007/s10096-014-2296-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/02/2014] [Indexed: 12/19/2022]
Abstract
This review summarizes evidence that commercially available fluoroquinolones used for the treatment of bacterial infections are active against other non-bacterial infectious agents as well. Any of these fluoroquinolones exerts, in parallel to its antibacterial action, antiviral, antifungal, and antiparasitic actions at clinically achievable concentrations. This broad range of anti-infective activities is due to one common mode of action, i.e., the inhibition of type II topoisomerases or inhibition of viral helicases, thus maintaining the selective toxicity of fluoroquinolones inhibiting microbial topoisomerases at low concentrations but mammalian topoisomerases at much higher concentrations. Evidence suggests that standard doses of the fluoroquinolones studied are clinically effective against viral and parasitic infections, whereas higher doses administered topically were active against Candida spp. causing ophthalmological infections. Well-designed clinical studies should be performed to substantiate these findings.
Collapse
Affiliation(s)
- A Dalhoff
- Institute for Infection Medicine, University Medical Center Schleswig-Holstein, Brunswiker Str. 4, 24105, Kiel, Germany,
| |
Collapse
|
20
|
Khan MA, Aljarbou AN, Khan A, Younus H. Liposomal thymoquinone effectively combats fluconazole-resistant Candida albicans in a murine model. Int J Biol Macromol 2015; 76:203-8. [PMID: 25709021 DOI: 10.1016/j.ijbiomac.2015.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/04/2015] [Accepted: 02/08/2015] [Indexed: 01/10/2023]
Abstract
The aim of the present study was to develop a novel liposomal formulation of thymoquinone (TQ) to treat fluconazole-susceptible and -resistant Candida albicans (C. albicans) infections. The liposomal preparation of TQ (Lip-TQ) was used against a fluconazole-susceptible or -resistant isolate of C. albicans. Various doses of fluconazole (0, 5, 10, 20 and 40 mg/kg) or free TQ or Lip-TQ (0, 1, 2 and 5mg/kg) were used to treat C. albicans infected mice. Mice were observed for 40 days post C. albicans infection, and their kidneys were assessed for the fungal load. Fluconazole showed anti-fungal activity against the drug-susceptible, but not against the -resistant isolate of C. albicans. Free TQ showed its activity against both fluconazole-susceptible or -resistant C. albicans, however, Lip-TQ was found to be the most effective and imparted ∼ 100% and ∼ 90% survival of mice infected with fluconazole-susceptible and -resistant isolates of C. albicans, respectively. Mice treated with Lip-TQ showed highly reduced severity of infection in their tissue homogenates. Therefore, Lip-TQ may effectively be used in the treatment of C. albicans infections, including those which are not responding to fluconazole.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Ahmad N Aljarbou
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
21
|
Zhang L, Chen Y, Wang L, Chen XP, Zhang WG, Wang CY, Wu HS. Chloroquine relieves acute lung injury in rats with acute hemorrhagic necrotizing pancreatitis. ACTA ACUST UNITED AC 2013; 33:357-360. [PMID: 23771660 PMCID: PMC7101714 DOI: 10.1007/s11596-013-1124-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Indexed: 12/15/2022]
Abstract
This study preliminarily investigated the mechanism by which chloroquine (CQ) relieves acute lung injury (ALI) complicated in acute hemorrhagic necrotizing pancreatitis (AHNP). Sixty male Wistar rats were randomized into sham-operated group (group A, n=10), AHNP group (group B, n=10), L-arginine-treated group (group C, n=10), L-N-nitro-L-arginine methyl ester (NAME)-treated group (group D, n=10), CQ-treated group (group E, n=10) and CQ+L-NAME-treated group (group F, n=10). TLR4 expression was measured by using real time-PCR and Western blotting respectively. The results showed that, in the group B, the expression of TLR4 and the levels of TNF-α and IL-6 in the lungs were significantly increased, and the nitric oxide (NO) concentration was reduced, as compared with those in the group A (P<0.05 or P<0.01). Lung injury was aggravated with the increased expression of TLR4. When the inhibitor and stimulator of TLR4, namely L-Arg and L-NAME, were added respectively, lung injury was correspondingly relieved or aggravated (P<0.05 or P<0.01). In the group E, TLR4 expression was substantially lower and NO concentration higher than those in the group B (P<0.05 or P<0.01). However, in the group F, NO concentration was markedly decreased, and the inhibitory effect of CQ on TLR4 expression and the relief of lung injury were weakened when compared with those in the group E (P<0.05 or P<0.01). It was concluded that TLR4 may play an important role in the pathogenesis and development of ALI complicated in AHNP. CQ could relieve ALI by decreasing the TLR4 expression and increasing the NO release.
Collapse
Affiliation(s)
- Lei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Chen
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Wang
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Ping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wan-Guang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chun-You Wang
- Center of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - He-Shui Wu
- Center of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
22
|
Shinde RB, Raut JS, Chauhan NM, Karuppayil SM. Chloroquine sensitizes biofilms of Candida albicans to antifungal azoles. Braz J Infect Dis 2013; 17:395-400. [PMID: 23602464 PMCID: PMC9428048 DOI: 10.1016/j.bjid.2012.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 11/18/2022] Open
Abstract
Biofilms formed by Candida albicans, a human pathogen, are known to be resistant to different antifungal agents. Novel strategies to combat the biofilm associated Candida infections like multiple drug therapy are being explored. In this study, potential of chloroquine to be a partner drug in combination with four antifungal agents, namely fluconazole, voriconazole, amphotericin B, and caspofungin, was explored against biofilms of C. albicans. Activity of various concentrations of chloroquine in combination with a particular antifungal drug was analyzed in a checkerboard format. Growth of biofilm in presence of drugs was analyzed by XTT-assay, in terms of relative metabolic activity compared to that of drug free control. Results obtained by XTT-metabolic assay were confirmed by scanning electron microscopy. The interactions between chloroquine and four antifungal drugs were determined by calculating fractional inhibitory concentration indices. Azole resistance in biofilms was reverted significantly (p < 0.05) in presence of 250 μg/mL of chloroquine, which resulted in inhibition of biofilms at very low concentrations of antifungal drugs. No significant alteration in the sensitivity of biofilms to caspofungin and amphotericin B was evident in combination with chloroquine. This study for the first time indicates that chloroquine potentiates anti-biofilm activity of fluconazole and voriconazole.
Collapse
|
23
|
Shinde RB, Rajput SB, Raut JS, Karuppayil SM. An in vitro repositioning study reveals antifungal potential of chloroquine to inhibit growth and morphogenesis in Candida albicans. J GEN APPL MICROBIOL 2013; 59:167-70. [DOI: 10.2323/jgam.59.167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
24
|
Abstract
INTRODUCTION Drug targeting to sites of tissue injury, tumor or infection with limited toxicity is the goal for successful pharmaceutics. Immunocytes (including mononuclear phagocytes (dendritic cells, monocytes and macrophages), neutrophils and lymphocytes) are highly mobile; they can migrate across impermeable barriers and release their drug cargo at sites of infection or tissue injury. Thus, immune cells can be exploited as Trojan horses for drug delivery. AREAS COVERED This paper reviews how immunocytes laden with drugs can cross the blood-brain or blood-tumor barriers to facilitate treatments for infectious diseases, injury, cancer, or inflammatory diseases. The promises and perils of cell-mediated drug delivery are reviewed, with examples of how immunocytes can be harnessed to improve therapeutic end points. EXPERT OPINION Using cells as delivery vehicles enables targeted drug transport and prolonged circulation times, along with reductions in cell and tissue toxicities. Such systems for drug carriage and targeted release represent a new disease-combating strategy being applied to a spectrum of human disorders. The design of nanocarriers for cell-mediated drug delivery may differ from those used for conventional drug delivery systems; nevertheless, engaging different defense mechanisms in drug delivery may open new perspectives for the active delivery of drugs.
Collapse
Affiliation(s)
- Elena V Batrakova
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| | | | | |
Collapse
|
25
|
Zhao Y, Haney MJ, Klyachko NL, Li S, Booth SL, Higginbotham SM, Jones J, Zimmerman MC, Mosley RL, Kabanov AV, Gendelman HE, Batrakova EV. Polyelectrolyte complex optimization for macrophage delivery of redox enzyme nanoparticles. Nanomedicine (Lond) 2011; 6:25-42. [PMID: 21182416 PMCID: PMC3037278 DOI: 10.2217/nnm.10.129] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND We posit that cell-mediated drug delivery can improve transport of therapeutic enzymes to the brain and decrease inflammation and neurodegeneration seen during Parkinson's disease. Our prior works demonstrated that macrophages loaded with nanoformulated catalase ('nanozyme') then parenterally injected protect the nigrostriatum in a murine model of Parkinson's disease. Packaging of catalase into block ionomer complex with a synthetic polyelectrolyte block copolymer precludes enzyme degradation in macrophages. METHODS We examined relationships between the composition and structure of block ionomer complexes with a range of block copolymers, their physicochemical characteristics, and loading, release and catalase enzymatic activity in bone marrow-derived macrophages. RESULTS Formation of block ionomer complexes resulted in improved aggregation stability. Block ionomer complexes with ε-polylysine and poly(L-glutamic acid)-poly(ethylene glycol) demonstrated the least cytotoxicity and high loading and release rates. However, these formulations did not efficiently protect catalase inside macrophages. CONCLUSION Nanozymes with polyethyleneimine- and poly(L-lysine)(10)-poly(ethylene glycol) provided the best protection of enzymatic activity for cell-mediated drug delivery.
Collapse
Affiliation(s)
- Yuling Zhao
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew J. Haney
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Natalia L. Klyachko
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Chemical Enzymology, Faculty of Chemistry, M.V.Lomonosov Moscow State University, Moscow, Russia
| | - Shu Li
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephanie L. Booth
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sheila M. Higginbotham
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jocelyn Jones
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew C. Zimmerman
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - R. Lee Mosley
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Center for Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Alexander V. Kabanov
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Howard E. Gendelman
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Center for Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Elena V. Batrakova
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
26
|
Beduneau A, Ma Z, Grotepas CB, Kabanov A, Rabinow BE, Gong N, Mosley RL, Dou H, Boska MD, Gendelman HE. Facilitated monocyte-macrophage uptake and tissue distribution of superparmagnetic iron-oxide nanoparticles. PLoS One 2009; 4:e4343. [PMID: 19183814 PMCID: PMC2629545 DOI: 10.1371/journal.pone.0004343] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2008] [Accepted: 12/28/2008] [Indexed: 12/02/2022] Open
Abstract
Background We posit that the same mononuclear phagocytes (MP) that serve as target cells and vehicles for a host of microbial infections can be used to improve diagnostics and drug delivery. We also theorize that physical and biological processes such as particle shape, size, coating and opsonization that affect MP clearance of debris and microbes can be harnessed to facilitate uptake of nanoparticles (NP) and tissue delivery. Methods Monocytes and monocyte-derived macrophages (MDM) were used as vehicles of superparamagnetic iron oxide (SPIO) NP and immunoglobulin (IgG) or albumin coated SPIO for studies of uptake and distribution. IgG coated SPIO was synthesized by covalent linkage and uptake into monocytes and MDM investigated related to size, time, temperature, concentration, and coatings. SPIO and IgG SPIO were infused intravenously into naïve mice. T2 measures using magnetic resonance imaging (MRI) were used to monitor tissue distribution in animals. Results Oxidation of dextran on the SPIO surface generated reactive aldehyde groups and permitted covalent linkage to amino groups of murine and human IgG and F(ab')2 fragments and for Alexa Fluor® 488 hydroxylamine to form a Schiff base. This labile intermediate was immediately reduced with sodium cyanoborohydride in order to stabilize the NP conjugate. Optical density measurements of the oxidized IgG, F(ab')2, and/or Alexa Fluor® 488 SPIO demonstrated ∼50% coupling yield. IgG-SPIO was found stable at 4°C for a period of 1 month during which size and polydispersity index varied little from 175 nm and 200 nm, respectively. In vitro, NP accumulated readily within monocyte and MDM cytoplasm after IgG-SPIO exposure; whereas, the uptake of native SPIO in monocytes and MDM was 10-fold less. No changes in cell viability were noted for the SPIO-containing monocytes and MDM. Cell morphology was not changed as observed by transmission electron microscopy. Compared to unconjugated SPIO, intravenous injection of IgG-SPIO afforded enhanced and sustained lymphoid tissue distribution over 24 hours as demonstrated by MRI. Conclusions Facilitated uptake of coated SPIO in monocytes and MDM was achieved. Uptake was linked to particle size and was time and concentration dependent. The ability of SPIO to be rapidly taken up and distributed into lymphoid tissues also demonstrates feasibility of macrophage-targeted nanoformulations for diagnostic and drug therapy.
Collapse
Affiliation(s)
- Arnaud Beduneau
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Zhiya Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Cassi B. Grotepas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Alexander Kabanov
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Barrett E. Rabinow
- Baxter Healthcare Corporation, Round Lake, Illinois, United States of America
| | - Nan Gong
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Huanyu Dou
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michael D. Boska
- Department of Radiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MDB); (HEG)
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MDB); (HEG)
| |
Collapse
|
27
|
Qiu L, Jing N, Jin Y. Preparation and in vitro evaluation of liposomal chloroquine diphosphate loaded by a transmembrane pH-gradient method. Int J Pharm 2008; 361:56-63. [PMID: 18573626 DOI: 10.1016/j.ijpharm.2008.05.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Revised: 04/30/2008] [Accepted: 05/12/2008] [Indexed: 11/27/2022]
Abstract
This study developed an active loading method for encapsulating chloroquine diphosphate (CQ) into liposomes. The effects of different formulation factors on the encapsulation efficiency (EE) and the size of CQ liposomes were investigated. These factors included the internal phase of liposomes, the external phase of liposomes, the ratio of drug to soybean phosphatidylcholine (drug/SPC), the ratio of cholesterol to soybean phosphatidylcholine (Chol/SPC), and the incubation temperature and time. The EE (93%) was obtained when using drug/SPC (1:50 mass ratio), SPC/Chol (1:5 mass ratio) at 0.10 M citrate-sodium citrate buffer (pH 3.6). As 5 mol% methoxypoly(ethylene glycol)(2,000) cholesteryl succinate (CHS-PEG(2000)) or distearoyl phosphatidylethanolamine-poly (ethylene glycol)(2,000) (DSPE-PEG(2000)) was added, the size of particle was reduced and the EE was improved. Freeze-drying with 5% trehalose as a cryoprotectant was carried out to achieve long-term stability. The drug release studies were performed in vitro simulating the desired application conditions, such as physiological fluids (pH 7.4), tumor tissues (pH 6.5) and endosomal compartments (pH 5.5). The release of CQ from the liposomes prepared via remote loading showed the significant pH-sensitivity and retention properties, which favored the application of liposomal CQ at tumor tissues and endosomal compartments.
Collapse
Affiliation(s)
- Liyan Qiu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | | | | |
Collapse
|
28
|
Alam MN, Ahmad FJ, Sultana Y, Iqbal Z, Aqil M, Ali A. Development and Characterization of Liposome‐Based Formulation of Amiloride Hydrochloride. J DISPER SCI TECHNOL 2008. [DOI: 10.1080/01932690701718735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
Briones E, Colino CI, Lanao JM. Delivery systems to increase the selectivity of antibiotics in phagocytic cells. J Control Release 2007; 125:210-27. [PMID: 18077047 DOI: 10.1016/j.jconrel.2007.10.027] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 10/29/2007] [Indexed: 11/25/2022]
Abstract
Many infectious diseases are caused by facultative organisms that are able to survive in phagocytic cells. The intracellular location of these microorganisms protects them from the host defence systems and from some antibiotics with poor penetration into phagocytic cells. One strategy used to improve the penetration of antibiotics into phagocytic cells is the use of carrier systems that deliver these drugs directly to the target cell. Delivery systems such as liposomes, micro/nanoparticles, lipid systems, conjugates, and biological carriers such as erythrocyte ghosts may contribute to increasing the therapeutic efficacy of antibiotics and antifungal agents in the treatment of infections caused by intracellular microorganisms. The main objective of this review is to analyze recent advances and current perspectives in the use of antibiotic delivery systems in the treatment of intracellular infections such as mycobacterial infections, brucellosis, salmonellosis, listeriosis, fungal infections, visceral leishmaniasis, and HIV.
Collapse
Affiliation(s)
- Elsa Briones
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain
| | | | | |
Collapse
|
30
|
Kechichian TB, Shea J, Del Poeta M. Depletion of alveolar macrophages decreases the dissemination of a glucosylceramide-deficient mutant of Cryptococcus neoformans in immunodeficient mice. Infect Immun 2007; 75:4792-8. [PMID: 17664261 PMCID: PMC2044542 DOI: 10.1128/iai.00587-07] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In previous studies we showed that a Cryptococcus neoformans mutant lacking glucosylceramide (Deltagcs1) is avirulent and unable to reach the brain when it is administered intranasally into an immunocompetent mouse and is contained in a lung granuloma. To determine whether granuloma formation is key for containment of C. neoformans Deltagcs1, we studied the role of C. neoformans glucosylceramide in a T- and NK-cell-immunodeficient mouse model (Tgepsilon26) in which alveolar macrophages (AMs) are not activated and granuloma formation is not expected. The results show that Tgepsilon26 mice infected with Deltagcs1 do not produce a lung granuloma and that the Deltagcs1 mutant proliferates in the lungs and does disseminate to the brain, although its virulence phenotype is dramatically reduced. Since Deltagcs1 can grow only in acidic niches, such as the phagolysosome of AMs, and not in neutral or alkaline environments, such as the extracellular spaces, we hypothesize that in immunodeficient mice Deltagcs1 proliferates inside AMs. Indeed, we found that depletion of AMs significantly improved Tgepsilon26 mouse survival and decreased the dissemination of Deltagcs1 cells to the central nervous system. Thus, these results suggest that the growth of Deltagcs1 in immunodeficient mice is maintained within AMs. This study highlights the hypothesis that AMs may exacerbate C. neoformans infection in conditions in which there is severe host immunodeficiency.
Collapse
Affiliation(s)
- Talar B Kechichian
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
31
|
Abstract
Neurodegenerative and infectious disorders including Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and stroke are rapidly increasing as population's age. Alzheimer's disease alone currently affects 4.5 million Americans, and more than $100 billion is spent per year on medical and institutional care for affected people. Such numbers will double in the ensuing decades. Currently disease diagnosis for all disorders is made, in large measure, on clinical grounds as laboratory and neuroimaging tests confirm what is seen by more routine examination. Achieving early diagnosis would enable improved disease outcomes. Drugs, vaccines or regenerative proteins present "real" possibilities for positively affecting disease outcomes, but are limited in that their entry into the brain is commonly restricted across the blood-brain barrier. This review highlights how these obstacles can be overcome by polymer science and nanotechnology. Such approaches may improve diagnostic and therapeutic outcomes. New developments in polymer science coupled with cell-based delivery strategies support the notion that diseases that now have limited therapeutic options can show improved outcomes by advances in nanomedicine.
Collapse
Affiliation(s)
- A.V. Kabanov
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - H.E. Gendelman
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
32
|
Khan MA, Khan A, Owais M. Prophylactic use of liposomized tuftsin enhances the susceptibility ofCandida albicansto fluconazole in leukopenic mice. ACTA ACUST UNITED AC 2006; 46:63-9. [PMID: 16420598 DOI: 10.1111/j.1574-695x.2005.00014.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the present study, we investigated the immunopotentiating activity of the immunomodulator tuftsin for the treatment of dose-dependent susceptible Candida albicans infection in a murine model. Our results demonstrated that tuftsin increases the susceptibility of C. albicans to phagocytosis by activating murine macrophages. Fluconazole used for the treatment of mice infected with C. albicans showed less in vivo efficacy and proved to be ineffective in the elimination of the infection from leukopenic mice even at higher doses. However, the antifungal activity of fluconazole against the same isolate of C. albicans significantly increased in tuftsin-pretreated mice and resulted in an improved survival rate in mice. The treated mice also showed less severity of infection as supported by a reduced fungal burden in their kidneys. This study indicates that the use of immunopotentiating substances can enhance the therapeutic efficacy of azole antifungal agents and thus can effectively combat azole-resistant fungal pathogens under conditions of immunosuppression.
Collapse
Affiliation(s)
- Masood A Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh-202002, India
| | | | | |
Collapse
|