1
|
Shapira T, Christofferson M, Av-Gay Y. The antimicrobial activity of innate host-directed therapies: A systematic review. Int J Antimicrob Agents 2024; 63:107138. [PMID: 38490573 DOI: 10.1016/j.ijantimicag.2024.107138] [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: 10/01/2023] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Intracellular human pathogens are the deadliest infectious diseases and are difficult to treat effectively due to their protection inside the host cell and the development of antimicrobial resistance (AMR). An emerging approach to combat these intracellular pathogens is host-directed therapies (HDT), which harness the innate immunity of host cells. HDT rely on small molecules to promote host protection mechanisms that ultimately lead to pathogen clearance. These therapies are hypothesized to: (1) possess indirect yet broad, cross-species antimicrobial activity, (2) effectively target drug-resistant pathogens, (3) carry a reduced susceptibility to the development of AMR and (4) have synergistic action with conventional antimicrobials. As the field of HDT expands, this systematic review was conducted to collect a compendium of HDT and their characteristics, such as the host mechanisms affected, the pathogen inhibited, the concentrations investigated and the magnitude of pathogen inhibition. The evidential support for the main four HDT hypotheses was assessed and concluded that HDT demonstrate robust cross-species activity, are active against AMR pathogens, clinical isolates and laboratory-adapted pathogens. However, limited information exists to support the notion that HDT are synergistic with canonical antimicrobials and are less predisposed to AMR development.
Collapse
Affiliation(s)
- Tirosh Shapira
- Department of Medicine, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Christofferson
- Department of Microbiology and Immunology, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yossef Av-Gay
- Department of Medicine, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada; Department of Microbiology and Immunology, Division of Infectious Disease, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Wu Y, Li L, Ning Z, Li C, Yin Y, Chen K, Li L, Xu F, Gao J. Autophagy-modulating biomaterials: multifunctional weapons to promote tissue regeneration. Cell Commun Signal 2024; 22:124. [PMID: 38360732 PMCID: PMC10868121 DOI: 10.1186/s12964-023-01346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/29/2023] [Indexed: 02/17/2024] Open
Abstract
Autophagy is a self-renewal mechanism that maintains homeostasis and can promote tissue regeneration by regulating inflammation, reducing oxidative stress and promoting cell differentiation. The interaction between biomaterials and tissue cells significantly affects biomaterial-tissue integration and tissue regeneration. In recent years, it has been found that biomaterials can affect various processes related to tissue regeneration by regulating autophagy. The utilization of biomaterials in a controlled environment has become a prominent approach for enhancing the tissue regeneration capabilities. This involves the regulation of autophagy in diverse cell types implicated in tissue regeneration, encompassing the modulation of inflammatory responses, oxidative stress, cell differentiation, proliferation, migration, apoptosis, and extracellular matrix formation. In addition, biomaterials possess the potential to serve as carriers for drug delivery, enabling the regulation of autophagy by either activating or inhibiting its processes. This review summarizes the relationship between autophagy and tissue regeneration and discusses the role of biomaterial-based autophagy in tissue regeneration. In addition, recent advanced technologies used to design autophagy-modulating biomaterials are summarized, and rational design of biomaterials for providing controlled autophagy regulation via modification of the chemistry and surface of biomaterials and incorporation of cells and molecules is discussed. A better understanding of biomaterial-based autophagy and tissue regeneration, as well as the underlying molecular mechanisms, may lead to new possibilities for promoting tissue regeneration. Video Abstract.
Collapse
Affiliation(s)
- Yan Wu
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Luxin Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Zuojun Ning
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Changrong Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yongkui Yin
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Kaiyuan Chen
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Lu Li
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Fei Xu
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Cellular and molecular level host-pathogen interactions in Francisella tularensis: A microbial gene network study. Comput Biol Chem 2021; 96:107601. [PMID: 34801846 DOI: 10.1016/j.compbiolchem.2021.107601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/12/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023]
Abstract
Due to the high infectivity and fatal effect on human population, Francisella tularensis (F. tularensis) is classified as a potential biological warfare agent. The interaction between host and pathogen behind the successful establishment of F. tularensis infection within the human host is largely unknown. In our present work, we have studied the molecular level interactions between the host cellular components and F. tularensis genes to understand the interplay between the host and pathogen. Interestingly, we have identified the pathways associated with the pathogen offensive strategies that help in invasion of host defensive systems. The F. tularensis genes purL, katG, proS, rpoB and fusA have displayed high number of interactions with the host genes and thus play a crucial role in vital pathogen pathways. The pathways identified were involved in adaptation to different stress conditions within the host and might be crucial for designing new therapeutic interventions against tularemia.
Collapse
|
4
|
Tsao N, Chang YC, Hsieh SY, Li TC, Chiu CC, Yu HH, Hsu TC, Kuo CF. AR-12 Has a Bactericidal Activity and a Synergistic Effect with Gentamicin against Group A Streptococcus. Int J Mol Sci 2021; 22:ijms222111617. [PMID: 34769046 PMCID: PMC8583967 DOI: 10.3390/ijms222111617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 12/30/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus (GAS) is an important human pathogen that can cause severe invasive infection, such as necrotizing fasciitis and streptococcal toxic shock syndrome. The mortality rate of streptococcal toxic shock syndrome ranges from 20% to 50% in spite of antibiotics administration. AR-12, a pyrazole derivative, has been reported to inhibit the infection of viruses, intracellular bacteria, and fungi. In this report, we evaluated the bactericidal activities and mechanisms of AR-12 on GAS infection. Our in vitro results showed that AR-12 dose-dependently reduced the GAS growth, and 2.5 μg/mL of AR-12 significantly killed GAS within 2 h. AR-12 caused a remarkable reduction in nucleic acid and protein content of GAS. The expression of heat shock protein DnaK and streptococcal exotoxins was also inhibited by AR-12. Surveys of the GAS architecture by scanning electron microscopy revealed that AR-12-treated GAS displayed incomplete septa and micro-spherical structures protruding out of cell walls. Moreover, the combination of AR-12 and gentamicin had a synergistic antibacterial activity against GAS replication for both in vitro and in vivo infection. Taken together, these novel findings obtained in this study may provide a new therapeutic strategy for invasive GAS infection.
Collapse
Affiliation(s)
- Nina Tsao
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Ya-Chu Chang
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Sung-Yuan Hsieh
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu 300024, Taiwan;
| | - Tang-Chi Li
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Ching-Chen Chiu
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Hai-Han Yu
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Tzu-Ching Hsu
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Chih-Feng Kuo
- School of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
- Department of Nursing, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
- Correspondence: ; Tel.: +886-7-6151100 (ext. 7967)
| |
Collapse
|
5
|
Zahid MSH, Varma DM, Johnson MM, Landavazo A, Bachelder EM, Blough BE, Ainslie KM. Overcoming reduced antibiotic susceptibility in intracellular Salmonella enterica serovar Typhimurium using AR-12. FEMS Microbiol Lett 2021; 368:6293843. [PMID: 34089315 DOI: 10.1093/femsle/fnab062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/02/2021] [Indexed: 11/14/2022] Open
Abstract
Host-directed therapies (HDTs) could enhance the activity of traditional antibiotics. AR-12 is a promising HDT against intracellular pathogens including Salmonella enterica serovar Typhimurium, and has been shown to act through modulation of autophagy and the Akt kinase pathway. Since AR-12 does not inhibit the growth of planktonic bacteria but only works in conjunction with the infected host-cell, we hypothesized that AR-12 could enhance the activity of antibiotics in less-susceptible strains in the intracellular host environment. We found that repetitive passaging of S. typhimurium in macrophages in the absence of antibiotics led to a 4-fold reduction in their intracellular susceptibility to streptomycin (STR), but had no effect on the bacteria's sensitivity to AR-12. Moreover, when the host-passaged strains were treated with a combined therapy of AR-12 and STR, there was a significant reduction of intracellular bacterial burden compared to STR monotherapy. Additionally, co-treatment of macrophages infected with multi-drug resistant S. typhimurium with AR-12 and STR or ampicillin showed enhanced clearance of the intracellular bacteria. The drug combination did not elicit this effect on planktonic bacteria. Overall, AR-12 enhanced the clearance of less susceptible S. typhimurium in an intracellular environment.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Devika M Varma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Antonio Landavazo
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,School of Medicine, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| |
Collapse
|
6
|
Ferreira PMP, Sousa RWRD, Ferreira JRDO, Militão GCG, Bezerra DP. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol Res 2021; 168:105582. [PMID: 33775862 DOI: 10.1016/j.phrs.2021.105582] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/09/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are the most common drugs used to relieve acute and chronic inflammatory diseases. In this article, we present a review about the use of CQ and HCQ in antitumor therapies based on autophagy mechanisms. These molecules break/discontinue autophagosome-lysosome fusions in initial phases and enhance antiproliferative action of chemotherapeutics. Their sensitizing effects of chemotherapy when used as an adjuvant option in clinical trials against cancer. However, human related-MDR genes are also under risk to develop chemo or radioresistance because cancer cells have ability to throw 4-aminoquinolines out from digestive vacuoles well. Additionally, they also have antitumor mechanism unrelated to autophagy, including cell death from apoptosis and necroptosis and immunomodulatory/anti-inflammatory properties. However, the link between some anticancer mechanisms, clinical efficacy and pharmacological safety has not yet been fully defined.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil.
| | - Rayran Walter Ramos de Sousa
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil
| | | | | | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ-BA), 40296-710 Salvador, Brazil
| |
Collapse
|
7
|
Mohan M, Bhattacharya D. Host-directed Therapy: A New Arsenal to Come. Comb Chem High Throughput Screen 2021; 24:59-70. [PMID: 32723230 DOI: 10.2174/1386207323999200728115857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 11/22/2022]
Abstract
The emergence of drug-resistant strains among the variety of pathogens worsens the situation in today's scenario. In such a situation, a very heavy demand for developing the new antibiotics has arisen, but unfortunately, very limited success has been achieved in this arena till now. Infectious diseases usually make their impression in the form of severe pathology. Intracellular pathogens use the host's cell machinery for their survival. They alter the gene expression of several host's pathways and endorse to shut down the cell's innate defense pathway like apoptosis and autophagy. Intracellular pathogens are co-evolved with hosts and have a striking ability to manipulate the host's factors. They also mimic the host molecules and secrete them to prevent the host's proper immune response against them for their survival. Intracellular pathogens in chronic diseases create excessive inflammation. This excessive inflammation manifests in pathology. Host directed therapy could be alternative medicine in this situation; it targets the host factors, and abrogates the replication and persistence of pathogens inside the cell. It also provokes the anti-microbial immune response against the pathogen and reduces the exacerbation by enhancing the healing process to the site of pathology. HDT targets the host's factor involved in a certain pathway that ultimately targets the pathogen life cycle and helps in eradication of the pathogen. In such a scenario, HDT could also play a significant role in the treatment of drugsensitive as well with drug resistance strains because it targets the host's factors, which favors the pathogen survival inside the cell.
Collapse
Affiliation(s)
- Mradul Mohan
- National Institute of Malaria Research, New Delhi, India
| | - Debapriya Bhattacharya
- Center for Biotechnology, School of Pharmaceutical Sciences, SOA Deemed University, Bhubaneswar, Odisha, India
| |
Collapse
|
8
|
Reggio A, Buonomo V, Grumati P. Eating the unknown: Xenophagy and ER-phagy are cytoprotective defenses against pathogens. Exp Cell Res 2020; 396:112276. [PMID: 32918896 PMCID: PMC7480532 DOI: 10.1016/j.yexcr.2020.112276] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 01/01/2023]
Abstract
Autophagy is an evolutionary conserved catabolic process devoted to the removal of unnecessary and harmful cellular components. In its general form, autophagy governs cellular lifecycle through the formation of double membrane vesicles, termed autophagosomes, that enwrap and deliver unwanted intracellular components to lysosomes. In addition to this omniscient role, forms of selective autophagy, relying on specialized receptors for cargo recognition, exert fine-tuned control over cellular homeostasis. In this regard, xenophagy plays a pivotal role in restricting the replication of intracellular pathogens, thus acting as an ancient innate defense system against infections. Recently, selective autophagy of the endoplasmic reticulum (ER), more simply ER-phagy, has been uncovered as a critical mechanism governing ER network shape and function. Six ER-resident proteins have been characterized as ER-phagy receptors and their orchestrated function enables ER homeostasis and turnover overtime. Unfortunately, ER is also the preferred site for viral replication and several viruses hijack ER machinery for their needs. Thus, it is not surprising that some ER-phagy receptors can act to counteract viral replication and minimize the spread of infection throughout the organism. On the other hand, evolutionary pressure has armed pathogens with strategies to evade and subvert xenophagy and ER-phagy. Although ER-phagy biology is still in its infancy, the present review aims to summarize recent ER-phagy literature, with a special focus on its role in counteracting viral infections. Moreover, we aim to offer some hints for future targeted approaches to counteract host-pathogen interactions by modulating xenophagy and ER-phagy pathways.
Collapse
Affiliation(s)
- Alessio Reggio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Viviana Buonomo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy.
| |
Collapse
|
9
|
AR-12 Exhibits Direct and Host-Targeted Antibacterial Activity toward Mycobacterium abscessus. Antimicrob Agents Chemother 2020; 64:AAC.00236-20. [PMID: 32482678 PMCID: PMC7526805 DOI: 10.1128/aac.00236-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
Therapeutic options for Mycobacterium abscessus infections are extremely limited. New or repurposed drugs are needed. The anti-M. abscessus activity of AR-12 (OSU-03012), reported to express broad-spectrum antimicrobial effects, was investigated in vitro and in vivo Antimicrobial susceptibility testing was performed on 194 clinical isolates. Minimum bactericidal concentration and time-kill kinetics assays were conducted to distinguish the bactericidal versus bacteriostatic activity of AR-12. Synergy between AR-12 and five clinically important antibiotics was determined using a checkerboard synergy assay. The activity of AR-12 against intracellular M. abscessus residing within macrophage was also evaluated. Finally, the potency of AR-12 in vivo was determined in a neutropenic mouse model that mimics pulmonary M. abscessus infection. AR-12 exhibited high anti-M. abscessus activity in vitro, with an MIC50 of 4 mg/liter (8.7 μM) and an MIC90 of 8 mg/liter (17.4 μM) for both subsp. abscessus and subsp. massiliense AR-12 and amikacin exhibited comparable bactericidal activity against extracellular M. abscessus in culture. AR-12, however, exhibited significantly greater intracellular antibacterial activity than amikacin and caused a significant reduction in the bacterial load in the lungs of neutropenic mice infected with M. abscessus No antagonism between AR-12 and clarithromycin, amikacin, imipenem, cefoxitin, or tigecycline was evident. In conclusion, AR-12 is active against M. abscessus in vitro and in vivo and does not antagonize the most frequently used anti-M. abscessus drugs. As such, AR-12 is a potential candidate to include in novel strategies to treat M. abscessus infections.
Collapse
|
10
|
Gregory DJ, DeLoid GM, Salmon SL, Metzger DW, Kramnik I, Kobzik L. SON DNA-binding protein mediates macrophage autophagy and responses to intracellular infection. FEBS Lett 2020; 594:2782-2799. [PMID: 32484234 DOI: 10.1002/1873-3468.13851] [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: 05/03/2020] [Accepted: 05/11/2020] [Indexed: 12/09/2022]
Abstract
Intracellular pathogens affect diverse host cellular defence and metabolic pathways. Here, we used infection with Francisella tularensis to identify SON DNA-binding protein as a central determinant of macrophage activities. RNAi knockdown of SON increases survival of human macrophages following F. tularensis infection or inflammasome stimulation. SON is required for macrophage autophagy, interferon response factor 3 expression, type I interferon response and inflammasome-associated readouts. SON knockdown has gene- and stimulus-specific effects on inflammatory gene expression. SON is required for accurate splicing and expression of GBF1, a key mediator of cis-Golgi structure and function. Chemical GBF1 inhibition has similar effects to SON knockdown, suggesting that SON controls macrophage functions at least in part by controlling Golgi-associated processes.
Collapse
Affiliation(s)
- David J Gregory
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Pediatric Infectious Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Glen M DeLoid
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sharon L Salmon
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Dennis W Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, MA, USA
| | - Lester Kobzik
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
Varma DM, Zahid MSH, Bachelder EM, Ainslie KM. Formulation of host-targeted therapeutics against bacterial infections. Transl Res 2020; 220:98-113. [PMID: 32268128 PMCID: PMC10132281 DOI: 10.1016/j.trsl.2020.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/23/2022]
Abstract
The global burden of bacterial infections is rising due to increasing resistance to the majority of first-line antibiotics, rendering these drugs ineffective against several clinically important pathogens. Limited transport of antibiotics into cells compounds this problem for gram-negative bacteria that exhibit prominent intracellular lifecycles. Furthermore, poor bioavailability of antibiotics in infected tissues necessitates higher doses and longer treatment regimens to treat resistant infections. Although emerging antibiotics can combat these problems, resistance still may develop over time. Expanding knowledge of host-pathogen interactions has inspired research and development of host-directed therapies (HDTs). HDTs target host-cell machinery critical for bacterial pathogenesis to treat bacterial infections alone or as adjunctive treatment with traditional antibiotics. Unlike traditional antibiotics that directly affect bacteria, a majority of HDTs function by boosting the endogenous antimicrobial activity of cells and are consequently less prone to bacterial tolerance induced by selection pressure. Therefore, HDTs can be quite effective against intracellular cytosolic or vacuolar bacteria, which a majority of traditional antibiotics are unable to eradicate. However, in vivo therapeutic efficacy of HDTs is reliant on adequate bioavailability. Particle-based formulations demonstrate the potential to enable targeted drug delivery, enhance cellular uptake, and increase drug concentration in the host cell of HDTs. This review selected HDTs for clinically important pathogens, identifies formulation strategies that can improve their therapeutic efficacy and offers insights toward further development of HDTs for bacterial infections.
Collapse
Affiliation(s)
- Devika M Varma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
12
|
Chou YJ, Lin CC, Dzhagalov I, Chen NJ, Lin CH, Lin CC, Chen ST, Chen KH, Fu SL. Vaccine adjuvant activity of a TLR4-activating synthetic glycolipid by promoting autophagy. Sci Rep 2020; 10:8422. [PMID: 32439945 PMCID: PMC7242473 DOI: 10.1038/s41598-020-65422-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/20/2020] [Indexed: 11/09/2022] Open
Abstract
Toll-like receptors (TLRs) play crucial roles in host immune defenses. Recently, TLR-mediated autophagy is reported to promote immune responses via increasing antigen processing and presentation in antigen presenting cells. The present study examined whether the synthetic TLR4 activator (CCL-34) could induce autophagy to promote innate and adaptive immunity. In addition, the potential of CCL-34 as an immune adjuvant in vivo was also investigated. Our data using RAW264.7 cells and bone marrow-derived macrophages showed that CCL-34 induced autophagy through a TLR4-NF-κB pathway. The autophagy-related molecules (Nrf2, p62 and Beclin 1) were activated in RAW264.7 cells and bone marrow-derived macrophages under CCL-34 treatment. CCL-34-stimulated macrophages exhibited significant antigen-processing activity and induced the proliferation of antigen-specific CD4+T cells as well as the production of activated T cell-related cytokines, IL-2 and IFN-γ. Furthermore, CCL-34 immunization in mice induced infiltration of monocytes in the peritoneal cavity and elevation of antigen-specific IgG in the serum. CCL-34 treatment in vivo did not cause toxicity based on serum biochemical profiles. Notably, the antigen-specific responses induced by CCL-34 were attenuated by the autophagy inhibitor, 3-methyladenine. In summary, we demonstrated CCL-34 can induce autophagy to promote antigen-specific immune responses and act as an efficient adjuvant.
Collapse
Affiliation(s)
- Yi-Ju Chou
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan
| | - Ching-Cheng Lin
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Ivan Dzhagalov
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Szu-Ting Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Kuo-Hsin Chen
- Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan.
| | - Shu-Ling Fu
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan. .,Institute of Traditional Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.
| |
Collapse
|
13
|
Abdulrahman BA, Tahir W, Doh-Ura K, Gilch S, Schatzl HM. Combining autophagy stimulators and cellulose ethers for therapy against prion disease. Prion 2020; 13:185-196. [PMID: 31578923 PMCID: PMC6779372 DOI: 10.1080/19336896.2019.1670928] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Prion diseases are fatal transmissible neurodegenerative disorders that affect animals and humans. Prions are proteinaceous infectious particles consisting of a misfolded isoform of the cellular prion protein PrPC, termed PrPSc. PrPSc accumulates in infected neurons due to partial resistance to proteolytic digestion. Using compounds that interfere with the production of PrPSc or enhance its degradation cure prion infection in vitro, but most drugs failed when used to treat prion-infected rodents. In order to synergize the effect of anti-prion drugs, we combined drugs interfering with the generation of PrPSc with compounds inducing PrPSc degradation. Here, we tested autophagy stimulators (rapamycin or AR12) and cellulose ether compounds (TC-5RW or 60SH-50) either as single or combination treatment of mice infected with RML prions. Single drug treatments significantly extended the survival compared to the untreated group. As anticipated, also all the combination therapy groups showed extended survival compared to the untreated group, but no combination treatment showed superior effects to 60SH-50 or TC-5RW treatment alone. Unexpectedly, we later found that combining autophagy stimulator and cellulose ether treatment in cultured neuronal cells mitigated the pro-autophagic activity of AR12 and rapamycin, which can in part explain the in vivo results. Overall, we show that it is critical to exclude antagonizing drug effects when attempting combination therapy. In addition, we identified AR-12 as a pro-autophagic drug that significantly extends survival of prion-infected mice, has no adverse side effects on the animals used in this study, and can be useful in future studies.
Collapse
Affiliation(s)
- Basant A Abdulrahman
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary , Calgary , Alberta , Canada.,Calgary Prion Research Unit, University of Calgary , Calgary , Alberta , Canada.,Hotchkiss Brain Institute, University of Calgary , Calgary , Alberta , Canada.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University , Cairo , Egypt
| | - Waqas Tahir
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary , Calgary , Alberta , Canada.,Calgary Prion Research Unit, University of Calgary , Calgary , Alberta , Canada.,Hotchkiss Brain Institute, University of Calgary , Calgary , Alberta , Canada
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Sabine Gilch
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary , Calgary , Alberta , Canada.,Calgary Prion Research Unit, University of Calgary , Calgary , Alberta , Canada.,Hotchkiss Brain Institute, University of Calgary , Calgary , Alberta , Canada
| | - Hermann M Schatzl
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary , Calgary , Alberta , Canada.,Calgary Prion Research Unit, University of Calgary , Calgary , Alberta , Canada.,Hotchkiss Brain Institute, University of Calgary , Calgary , Alberta , Canada
| |
Collapse
|
14
|
Zahid MSH, Johnson MM, Tokarski RJ, Satoskar AR, Fuchs JR, Bachelder EM, Ainslie KM. Evaluation of synergy between host and pathogen-directed therapies against intracellular Leishmania donovani. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 10:125-132. [PMID: 31493763 PMCID: PMC6731340 DOI: 10.1016/j.ijpddr.2019.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 11/24/2022]
Abstract
Visceral leishmaniasis (VL) is associated with treatment complications due to the continued growth of resistant parasites toward currently available pathogen-directed therapeutics. To limit the emergence and combat resistant parasites there is a need to develop new anti-leishmanial drugs and alternative treatment approaches, such as host-directed therapeutics (HDTs). Discovery of new anti-leishmanial drugs including HDTs requires suitable in vitro assay systems. Herein, we modified and evaluated a series of resazurin assays against different life-stages of the VL causing parasite, Leishmania donovani to identify novel HDTs. We further analyzed the synergy of combinatorial interactions between traditionally used pathogen-directed drugs and HDTs for clearance of intracellular L. donovani. The inhibitory concentration at 50% (IC50) of the five evaluated therapies [amphotericin B (AMB), miltefosine, paromomycin, DNER-4, and AR-12 (OSU-03012)] was determined against promastigotes, extracellular amastigotes, and intracellular amastigotes of L. donovani via a resazurin-based assay and compared to image-based microscopy. Using the resazurin-based assay, all evaluated therapies showed reproducible anti-leishmanial activity against the parasite's different life-stages. These results were consistent to the traditional image-based technique. The gold standard of therapy, AMB, showed the highest potency against intracellular L. donovani, and was further evaluated for combinatorial effects with the HDTs. Among the combinations analyzed, pathogen-directed AMB and host-directed AR-12 showed a synergistic reduction of intracellular L. donovani compared to individual treatments. The modified resazurin assay used in this study demonstrated a useful technique to measure new anti-leishmanial drugs against both intracellular and extracellular parasites. The synergistic interactions between pathogen-directed AMB and host-directed AR-12 showed a great promise to combat VL, with the potential to reduce the emergence of drug-resistant strains.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert J Tokarski
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Abhay R Satoskar
- Department of Pathology, Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
15
|
Yang CY, Hsu CY, Fang CS, Shiau CW, Chen CS, Chiu HC. Loxapine, an antipsychotic drug, suppresses intracellular multiple-antibiotic-resistant Salmonella enterica serovar Typhimurium in macrophages. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:638-647. [DOI: 10.1016/j.jmii.2019.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/11/2019] [Accepted: 05/29/2019] [Indexed: 11/28/2022]
|
16
|
Khandia R, Dadar M, Munjal A, Dhama K, Karthik K, Tiwari R, Yatoo MI, Iqbal HMN, Singh KP, Joshi SK, Chaicumpa W. A Comprehensive Review of Autophagy and Its Various Roles in Infectious, Non-Infectious, and Lifestyle Diseases: Current Knowledge and Prospects for Disease Prevention, Novel Drug Design, and Therapy. Cells 2019; 8:cells8070674. [PMID: 31277291 PMCID: PMC6678135 DOI: 10.3390/cells8070674] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 02/05/2023] Open
Abstract
Autophagy (self-eating) is a conserved cellular degradation process that plays important roles in maintaining homeostasis and preventing nutritional, metabolic, and infection-mediated stresses. Autophagy dysfunction can have various pathological consequences, including tumor progression, pathogen hyper-virulence, and neurodegeneration. This review describes the mechanisms of autophagy and its associations with other cell death mechanisms, including apoptosis, necrosis, necroptosis, and autosis. Autophagy has both positive and negative roles in infection, cancer, neural development, metabolism, cardiovascular health, immunity, and iron homeostasis. Genetic defects in autophagy can have pathological consequences, such as static childhood encephalopathy with neurodegeneration in adulthood, Crohn's disease, hereditary spastic paraparesis, Danon disease, X-linked myopathy with excessive autophagy, and sporadic inclusion body myositis. Further studies on the process of autophagy in different microbial infections could help to design and develop novel therapeutic strategies against important pathogenic microbes. This review on the progress and prospects of autophagy research describes various activators and suppressors, which could be used to design novel intervention strategies against numerous diseases and develop therapeutic drugs to protect human and animal health.
Collapse
Affiliation(s)
- Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal 462 026, Madhya Pradesh, India
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj 31975/148, Iran
| | - Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal 462 026, Madhya Pradesh, India.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India.
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai, Tamil Nadu 600051, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura, Uttar Pradesh 281 001, India
| | - Mohd Iqbal Yatoo
- Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Srinagar 190025, Jammu and Kashmir, India
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N. L., CP 64849, Mexico
| | - Karam Pal Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India
| | - Sunil K Joshi
- Department of Pediatrics, Division of Hematology, Oncology and Bone Marrow Transplantation, University of Miami School of Medicine, Miami, FL 33136, USA.
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
17
|
Krishnamurthy M, Lemmon MM, Falcinelli EM, Sandy RA, Dootz JN, Mott TM, Rajamani S, Schaecher KE, Duplantier AJ, Panchal RG. Enhancing the antibacterial activity of polymyxins using a nonantibiotic drug. Infect Drug Resist 2019; 12:1393-1405. [PMID: 31239720 PMCID: PMC6555264 DOI: 10.2147/idr.s196874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/06/2019] [Indexed: 11/24/2022] Open
Abstract
Purpose: The rapid emergence of multidrug-resistant (MDR) bacteria and the lack of new therapies to eliminate them poses a major threat to global health. With the alarming rise in antimicrobial resistance (AMR), focus has now shifted to the use of the polymyxin class of antibiotics as the last line of defense for treatment of Gram-negative infections. Unfortunately, the growing resistance of bacteria against polymyxins is threatening the treatment of MDR infections, necessitating the need for novel strategies. The objective of this study was to determine if combination of polymyxin (polymyxin B or colistin) with a nonantibiotic small molecule AR-12, a celecoxib derivative that is devoid of cyclooxygenase 2 (COX-2) inhibitory activities, can be an effective strategy against polymyxin-resistant MDR bacteria. Methods: Growth inhibition studies, time-kill assays and permeability assays were conducted to investigate the effect of AR-12 on the antibacterial activity of polymyxins. Results: Growth studies were performed on a panel of polymyxin-resistant MDR strains using the combination of AR-12 with either colistin or polymyxin B. The combination treatment had no effect on strains that have inherent polymyxin resistance; however, AR-12 was effective in lowering the minimal inhibitory concentration (MIC) of polymyxins by 4–60-fold in several strains that had acquired polymyxin resistance. Time-kill assays using the combination of AR-12 and colistin with select MDR strains suggest rapid killing and bactericidal activity, while the permeability assays using fluorescently labeled dansylated polymyxin and 1-N-phenylnaphthylamine (NPN) in these MDR strains suggest that AR-12 can potentiate the antibacterial activity of polymyxins by possibly altering the bacterial outer membrane via modification of lipopolysaccharide and thereby improving the uptake of polymyxins. Conclusion: Our studies indicate that the combination of AR-12 and polymyxin is effective in targeting select Gram-negative bacteria that have acquired polymyxin resistance. Further understanding of the mechanism of action of AR-12 will provide new avenues for developing narrow-spectrum antibacterials to target select Gram-negative MDR bacteria. Importantly, our studies show that the use of nonantibiotic small molecules in combination with polymyxins is an attractive strategy to counter the growing resistance of bacteria to polymyxins.
Collapse
Affiliation(s)
- Malathy Krishnamurthy
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Margaret M Lemmon
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Evan M Falcinelli
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Reuel A Sandy
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Jennifer N Dootz
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Tiffany M Mott
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Sathish Rajamani
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA.,General Dynamics Information Technology, Frederick, MD, USA
| | - Kurt E Schaecher
- Department of Pathology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Allen J Duplantier
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA.,Cherokee Nation Assurance, Frederick, MD, USA
| | - Rekha G Panchal
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| |
Collapse
|
18
|
Novel AR-12 derivatives, P12-23 and P12-34, inhibit flavivirus replication by blocking host de novo pyrimidine biosynthesis. Emerg Microbes Infect 2018; 7:187. [PMID: 30459406 PMCID: PMC6246607 DOI: 10.1038/s41426-018-0191-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/12/2018] [Accepted: 10/21/2018] [Indexed: 12/14/2022]
Abstract
The genus Flavivirus contains many important pathogens, including dengue virus (DENV), Zika virus (ZIKV), and Japanese encephalitis virus (JEV). AR-12 is a celecoxib-derived anticancer agent that possesses antiviral activity against a broad range of viruses. We pharmacologically exploited this unique activity to develop additional antiviral agents, resulting in the production of the AR-12 derivatives P12-23 and P12-34. At nanomolar concentrations, these compounds were effective in suppressing DENV, ZIKV and JEV replication, exhibiting 10-fold improvements in the efficacy and selectivity indices as compared to AR-12. Regarding the mode of antiviral action, P12-23 and P12-34 inhibited viral RNA replication but had no effect on viral binding, entry or translation. Moreover, these AR-12 derivatives co-localized with mitochondrial markers, and their antiviral activity was lost in mitochondria-depleted cells. Interestingly, exogenous uridine or orotate, the latter being a metabolite of the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH), abolished the antiviral activity of AR-12 and its derivatives. As DHODH is a key enzyme in the de novo pyrimidine biosynthesis pathway, these AR-12 derivatives may act by targeting pyrimidine biosynthesis in host cells to inhibit viral replication. Importantly, treatment with P12-34 significantly improved the survival of mice that were subcutaneously challenged with DENV. Thus, P12-34 may warrant further evaluation as a therapeutic to control flaviviral outbreaks.
Collapse
|
19
|
Johnson MM, Collier MA, Hoang KV, Pino EN, Gurysh EG, Gallovic MD, Zahid MSH, Chen N, Schlesinger LS, Gunn JS, Bachelder EM, Ainslie KM. In Vivo and Cellular Trafficking of Acetalated Dextran Microparticles for Delivery of a Host-Directed Therapy for Salmonella enterica Serovar Typhi Infection. Mol Pharm 2018; 15:5336-5348. [PMID: 30296381 PMCID: PMC6330710 DOI: 10.1021/acs.molpharmaceut.8b00802] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previously we have encapsulated host-directed therapy AR-12 into acetalated dextran (Ace-DEX) microparticles (MPs) to mitigate drug toxicity and passively target phagocytic host cells. Herein, we have improved upon our initial emulsion-based formulation of Ace-DEX MPs encapsulating AR-12 (AR-12/MPs) by improving the drug encapsulation efficiency, evaluating sterilization processes for manufacturing, and understanding cellular and in vivo trafficking of the MPs. By using an alternative solvent system, ethyl acetate, we report an increased encapsulation efficiency of AR-12 while maintaining the pH-responsive degradation kinetics of Ace-DEX MPs. To better manufacture this novel antimicrobial formulation, we sterilized AR-12/MPs by gamma irradiation or ethylene oxide and evaluated their efficacy against intracellular Salmonella enterica serovar Typhi. Sterilized AR-12/MPs resulted in a significant reduction in intracellular bacterial burden compared to Blank/MPs. We also characterized intracellular trafficking of Ace-DEX MPs encapsulating fluorophores, which demonstrated internalization of MPs in endo/lysosomal compartments and time and degradation-rate dependent lysosomal escape into cytosolic compartments. Additionally, in vivo toxicity was mitigated following encapsulation of AR-12, where the maximum tolerated dose of AR-12 was increased compared to soluble treatment via intranasal, intravenous, and intraperitoneal administration routes. Following in vivo trafficking of Ace-DEX MPs via the same routes, intranasal administration demonstrated the highest accumulation in the lungs, liver, and kidneys, which persisted out to 240 h. Overall, we have advanced the formulation of this host-directed therapy and broadened the understanding of Ace-DEX MP delivery.
Collapse
Affiliation(s)
- Monica M. Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael A. Collier
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ky V. Hoang
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Erica N. Pino
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth G. Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew D. Gallovic
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Md. Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Naihan Chen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - John S. Gunn
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
20
|
Assani K, Shrestha CL, Rinehardt H, Zhang S, Robledo-Avila F, Wellmerling J, Partida-Sanchez S, Cormet-Boyaka E, Reynolds SD, Schlesinger LS, Kopp BT. AR-13 reduces antibiotic-resistant bacterial burden in cystic fibrosis phagocytes and improves cystic fibrosis transmembrane conductance regulator function. J Cyst Fibros 2018; 18:622-629. [PMID: 30366849 DOI: 10.1016/j.jcf.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 09/13/2018] [Accepted: 10/14/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND There are no effective treatments for Burkholderia cenocepacia in patients with cystic fibrosis (CF) due to bacterial multi-drug resistance and defective host killing. We demonstrated that decreased bacterial killing in CF is caused by reduced macrophage autophagy due to defective cystic fibrosis transmembrane conductance regulator (CFTR) function. AR-12 is a small molecule autophagy inducer that kills intracellular pathogens such as Francisella. We evaluated the efficacy of AR-12 and a new analogue AR-13 in reducing bacterial burden in CF phagocytes. METHODS Human CF and non-CF peripheral blood monocyte-derived macrophages, neutrophils, and nasal epithelial cells were exposed to CF bacterial strains in conjunction with treatment with antibiotics and/or AR compounds. RESULTS AR-13 and not AR-12 had growth inhibition on B. cenocepacia and methicillin-resistantStaphylococcus aureus (MRSA) in media alone. There was a 99% reduction in MRSA in CF macrophages, 71% reduction in Pseudomonas aeruginosa in CF neutrophils, and 70% reduction in non-CF neutrophils using AR-13. Conversely, there was no reduction in B. cenocepacia in infected CF and non-CF macrophages using AR-13 alone, but AR-13 and antibiotics synergistically reduced B. cenocepacia in CF macrophages. AR-13 improved autophagy in CF macrophages and CF patient-derived epithelial cells, and increased CFTR protein expression and channel function in CF epithelial cells. CONCLUSIONS The novel AR-12 analogue AR-13, in combination with antibiotics, reduced antibiotic-resistant bacterial burden in CF phagocytes, which correlated with increased autophagy and CFTR expression. AR-13 is a novel therapeutic for patients infected with B. cenocepacia and other resistant organisms that lack effective therapies.
Collapse
Affiliation(s)
- Kaivon Assani
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Chandra L Shrestha
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Hannah Rinehardt
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Shuzhong Zhang
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Frank Robledo-Avila
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Jack Wellmerling
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Susan D Reynolds
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | - Benjamin T Kopp
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, United States.
| |
Collapse
|
21
|
MicroRNA-27a controls the intracellular survival of Mycobacterium tuberculosis by regulating calcium-associated autophagy. Nat Commun 2018; 9:4295. [PMID: 30327467 PMCID: PMC6191460 DOI: 10.1038/s41467-018-06836-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/27/2018] [Indexed: 01/20/2023] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) kills millions every year, and there is urgent need to develop novel anti-TB agents due to the fast-growing of drug-resistant TB. Although autophagy regulates the intracellular survival of Mtb, the role of calcium (Ca2+) signaling in modulating autophagy during Mtb infection remains largely unknown. Here, we show that microRNA miR-27a is abundantly expressed in active TB patients, Mtb-infected mice and macrophages. The target of miR-27a is the ER-located Ca2+ transporter CACNA2D3. Targeting of this transporter leads to the downregulation of Ca2+ signaling, thus inhibiting autophagosome formation and promoting the intracellular survival of Mtb. Mice lacking of miR-27a and mice treated with an antagomir to miR-27a are more resistant to Mtb infection. Our findings reveal a strategy for Mtb to increase intracellular survival by manipulating the Ca2+-associated autophagy, and may also support the development of host-directed anti-TB therapeutic approaches. How Mycobacterium tuberculosis (Mtb) escapes autophagy-mediated clearance is poorly understood. Here, Liu et al. show that Mtb-induced MicroRNA-27a targets the ER-associated calcium transporter CACNA2D3, leading to suppression of antimicrobial autophagy and to enhanced intracellular survival of Mtb.
Collapse
|
22
|
The celecoxib derivative kinase inhibitor AR-12 (OSU-03012) inhibits Zika virus via down-regulation of the PI3K/Akt pathway and protects Zika virus-infected A129 mice: A host-targeting treatment strategy. Antiviral Res 2018; 160:38-47. [PMID: 30326204 PMCID: PMC7113887 DOI: 10.1016/j.antiviral.2018.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022]
Abstract
Zika virus (ZIKV) is a human-pathogenic flavivirus that has recently emerged as a global public health threat. ZIKV infection may be associated with congenital malformations in infected fetuses and severe neurological and systemic complications in infected adults. There are currently limited treatment options for ZIKV infection. AR-12 (OSU-03012) is a celecoxib derivative cellular kinase inhibitor that has broad-spectrum antiviral activities. In this study, we investigated the antiviral activity and mechanism of AR-12 against ZIKV. We evaluated the in vitro anti-ZIKV activity of AR-12, using cell protection and virus yield reduction assays, in multiple clinically relevant cell lines, and the in vivo treatment effects of AR-12 in a lethal mouse model using type I interferon receptor-deficient A129 mice. AR-12 inhibited ZIKV strains belonging to both the African and Asian/American lineages in Huh-7 and/or neuronal cells. AR12's IC50 against ZIKV was consistently <2 μM in these cells. ZIKV-infected A129 mice treated with intraperitoneally or orally administered AR-12 had significantly higher survival rate (50.0%–83.3% vs 0%, P < 0.05), less body weight loss, and lower blood and tissue ZIKV RNA loads than untreated control A129 mice. These anti-ZIKV effects were likely the results of down-regulation of the PI3K/Akt pathway by AR-12. Clinical trials using the clinically available and broad-spectrum AR-12 as an empirical treatment should be considered especially for patients residing in or returning from areas endemic of ZIKV and other arboviral infections who present with an acute undifferentiated febrile illness. AR-12 (OSU-03012) inhibited the replication of Zika virus strains belonging to both the Asian/American and African lineages. AR-12 inhibited Zika virus replication in multiple cell types in vitro. AR-12 treatment improved clinical and virological outcome of Zika virus-infected type I interferon receptor-deficient mice. AR-12 inhibited Zika virus replication via down-regulation of protein kinase B (Akt).
Collapse
|
23
|
Zhao X, Tang X, Guo N, An Y, Chen X, Shi C, Wang C, Li Y, Li S, Xu H, Liu M, Wang Y, Yu L. Biochanin a Enhances the Defense Against Salmonella enterica Infection Through AMPK/ULK1/mTOR-Mediated Autophagy and Extracellular Traps and Reversing SPI-1-Dependent Macrophage (MΦ) M2 Polarization. Front Cell Infect Microbiol 2018; 8:318. [PMID: 30271755 PMCID: PMC6142880 DOI: 10.3389/fcimb.2018.00318] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/21/2018] [Indexed: 12/15/2022] Open
Abstract
A novel treatment regimen for bacterial infections is the pharmacological enhancement of the host's immune defenses. We demonstrated that biochanin A (BCA), an isoflavone constituent in some plants, could enhance both intra- and extracellular bactericidal activity of host cells. First, BCA could induce a complete autophagic response in nonphagocytic cells (HeLa) or macrophages (MΦ) via the AMPK/ULK1/mTOR pathway and Beclin-1-dependent manner, and BCA enhanced the killing of invading Salmonella by autophagy through reinforcing ubiquitinated adapter protein (LRSAM1, NDP52 and p62)-mediated recognition of intracellular bacteria and through the formation of autophagolysosomes. Second, we demonstrated that BCA could enhance the release of MΦ extracellular traps (METs) to remove extracellular Salmonella also via the AMPK/ULK1/mTOR pathway, not through reactive oxygen species (ROS) pathway. Furtherly, in a Salmonella-infected mouse model, BCA treatment increased intra- and extracellular bactericidal activity through the strengthening autophagy and MET production, respectively, in peritoneal MΦ, liver and spleen tissue. Additionally, our findings showed that BCA downregulated SPI-1 (Salmonella pathogenicity island 1) expression during Salmonella infection in vitro and in vivo to reverse the MΦ M2 polarization, which was different from the MΦ M1 phenotype caused by most of bacteria infection. Together, these findings suggest that BCA has an immunomodulatory effect on Salmonella-infected host cells and enhances their bactericidal activity in vitro and in vivo through autophagy, extracellular traps and regulation of MΦ polarization.
Collapse
Affiliation(s)
- Xingchen Zhao
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China.,Department of Food Quality and Safety, College of Food Science and Engineering, Tonghua Normal University, Tonghua, China
| | - Xudong Tang
- Key Lab for New Drug Research of TCM, Research Institute of Tsinghua University in Shenzhen, Shenzhen, China
| | - Na Guo
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yanan An
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiangrong Chen
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Ce Shi
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Chao Wang
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yan Li
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Shulin Li
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Hongyue Xu
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Mingyuan Liu
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yang Wang
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lu Yu
- Key Laboratory for Zoonosis Research, Department of Infectious Diseases, First Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine, College of Food Science and Engineering, Institute of Zoonosis, Jilin University, Changchun, China
| |
Collapse
|
24
|
Clemens DL, Lee BY, Horwitz MA. The Francisella Type VI Secretion System. Front Cell Infect Microbiol 2018; 8:121. [PMID: 29740542 PMCID: PMC5924787 DOI: 10.3389/fcimb.2018.00121] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/03/2018] [Indexed: 12/27/2022] Open
Abstract
Francisella tularensisis subsp. tularensis is an intracellular bacterial pathogen and the causative agent of the life-threatening zoonotic disease tularemia. The Francisella Pathogenicity Island encodes a large secretion apparatus, known as a Type VI Secretion System (T6SS), which is essential for Francisella to escape from its phagosome and multiply within host macrophages and to cause disease in animals. The T6SS, found in one-quarter of Gram-negative bacteria including many highly pathogenic ones, is a recently discovered secretion system that is not yet fully understood. Nevertheless, there have been remarkable advances in our understanding of the structure, composition, and function of T6SSs of several bacteria in the past few years. The system operates like an inside-out headless contractile phage that is anchored to the bacterial membrane via a baseplate and membrane complex. The system injects effector molecules across the inner and outer bacterial membrane and into host prokaryotic or eukaryotic targets to kill, intoxicate, or in the case of Francisella, hijack the target cell. Recent advances include an atomic model of the contractile sheath, insights into the mechanics of sheath contraction, the composition of the baseplate and membrane complex, the process of assembly of the apparatus, and identification of numerous effector molecules and activities. While Francisella T6SS appears to be an outlier among T6SSs, with limited or no sequence homology with other systems, its structure and organization are strikingly similar to other systems. Nevertheless, we have only scratched the surface in uncovering the mysteries of the Francisella T6SS, and there are numerous questions that remain to be answered.
Collapse
Affiliation(s)
- Daniel L. Clemens
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
25
|
Mitigating the Impact of Antibacterial Drug Resistance through Host-Directed Therapies: Current Progress, Outlook, and Challenges. mBio 2018; 9:mBio.01932-17. [PMID: 29382729 PMCID: PMC5790911 DOI: 10.1128/mbio.01932-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increasing incidences of multidrug resistance in pathogenic bacteria threaten our ability to treat and manage bacterial infection. The development and FDA approval of novel antibiotics have slowed over the past decade; therefore, the adoption and improvement of alternative therapeutic strategies are critical for addressing the threat posed by multidrug-resistant bacteria. Host-directed therapies utilize small-molecule drugs and proteins to alter the host response to pathogen infection. Here, we highlight strategies for modulating the host inflammatory response to enhance bacterial clearance, small-molecule potentiation of innate immunity, and targeting of host factors that are exploited by pathogen virulence factors. Application of state-of-the-art "omic" technologies, including proteomics, transcriptomics, and image-omics (image-based high-throughput phenotypic screening), combined with powerful bioinformatics tools will enable the modeling of key signaling pathways in the host-pathogen interplay and aid in the identification of host proteins for therapeutic targeting and the discovery of host-directed small molecules that will regulate bacterial infection. We conclude with an outlook on research needed to overcome the challenges associated with transitioning host-directed therapies into a clinical setting.
Collapse
|
26
|
Gregory DJ, Kramnik I, Kobzik L. Protection of macrophages from intracellular pathogens by miR-182-5p mimic-a gene expression meta-analysis approach. FEBS J 2017; 285:244-260. [PMID: 29197182 DOI: 10.1111/febs.14348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/29/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
The goals of this study were to (a) define which host genes are of particular importance during the interactions between macrophages and intracellular pathogens, and (b) use this knowledge to gain fresh, experimental understanding of how macrophage activities may be manipulated during host defense. We designed an in silico method for meta-analysis of microarray gene expression data, and used this to combine data from 16 different studies of cells in the monocyte-macrophage lineage infected with seven different pathogens. Three thousand four hundred ninety-eight genes were identified, which we call the macrophage intracellular pathogen response (macIPR) gene set. As expected, the macIPR gene set showed a strong bias toward genes previously associated with the immune response. Predicted target sites for miR-182-5p (miR-182) were strongly over-represented among macIPR genes, indicating an unexpected role for miR-182-regulatable genes during intracellular pathogenesis. We therefore transfected primary human alveolar macrophage-like monocyte-derived macrophages from multiple different donors with synthetic miR-182, and found that miR-182 overexpression (a) increases proinflammatory gene induction during infection with Francisella tularensis live vaccine strain (LVS), (b) primes macrophages for increased autophagy, and (c) enhances macrophage control of both gram negative F. tularensisLVS and gram positive Bacillus anthracisANR-1 spores. These data therefore suggest a new application for miR-182 in promoting resistance to intracellular pathogens.
Collapse
Affiliation(s)
- David J Gregory
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, MA, USA
| | - Lester Kobzik
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
27
|
Abdulrahman BA, Abdelaziz D, Thapa S, Lu L, Jain S, Gilch S, Proniuk S, Zukiwski A, Schatzl HM. The celecoxib derivatives AR-12 and AR-14 induce autophagy and clear prion-infected cells from prions. Sci Rep 2017; 7:17565. [PMID: 29242534 PMCID: PMC5730578 DOI: 10.1038/s41598-017-17770-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/30/2017] [Indexed: 01/05/2023] Open
Abstract
Prion diseases are fatal infectious neurodegenerative disorders that affect both humans and animals. The autocatalytic conversion of the cellular prion protein (PrPC) into the pathologic isoform PrPSc is a key feature in prion pathogenesis. AR-12 is an IND-approved derivative of celecoxib that demonstrated preclinical activity against several microbial diseases. Recently, AR-12 has been shown to facilitate clearance of misfolded proteins. The latter proposes AR-12 to be a potential therapeutic agent for neurodegenerative disorders. In this study, we investigated the role of AR-12 and its derivatives in controlling prion infection. We tested AR-12 in prion infected neuronal and non-neuronal cell lines. Immunoblotting and confocal microscopy results showed that AR-12 and its analogue AR-14 reduced PrPSc levels after only 72 hours of treatment. Furthermore, infected cells were cured of PrPSc after exposure of AR-12 or AR-14 for only two weeks. We partially attribute the influence of the AR compounds on prion propagation to autophagy stimulation, in line with our previous findings that drug-induced stimulation of autophagy has anti-prion effects in vitro and in vivo. Taken together, this study demonstrates that AR-12 and the AR-14 analogue are potential new therapeutic agents for prion diseases and possibly protein misfolding disorders involving prion-like mechanisms.
Collapse
Affiliation(s)
- Basant A Abdulrahman
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy, Helwan University, 11795, Cairo, Egypt
| | - Dalia Abdelaziz
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy, Helwan University, 11795, Cairo, Egypt
| | - Simrika Thapa
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
| | - Li Lu
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
| | - Shubha Jain
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
| | - Sabine Gilch
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Department of Ecosystem & Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada
| | | | | | - Hermann M Schatzl
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada.
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, T2N 4Z6, Canada.
- Departments of Veterinary Sciences and of Molecular Biology, University of Wyoming, Laramie, Wyoming, 82071, USA.
| |
Collapse
|
28
|
Losada-Echeberría M, Herranz-López M, Micol V, Barrajón-Catalán E. Polyphenols as Promising Drugs against Main Breast Cancer Signatures. Antioxidants (Basel) 2017; 6:E88. [PMID: 29112149 PMCID: PMC5745498 DOI: 10.3390/antiox6040088] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/30/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is one of the most common neoplasms worldwide, and in spite of clinical and pharmacological advances, it is still a clinical problem, causing morbidity and mortality. On the one hand, breast cancer shares with other neoplasms some molecular signatures such as an imbalanced redox state, cell cycle alterations, increased proliferation and an inflammatory status. On the other hand, breast cancer shows differential molecular subtypes that determine its prognosis and treatment. These are characterized mainly by hormone receptors especially estrogen receptors (ERs) and epidermal growth factor receptor 2 (HER2). Tumors with none of these receptors are classified as triple negative breast cancer (TNBC) and are associated with a worse prognosis. The success of treatments partially depends on their specificity and the adequate molecular classification of tumors. New advances in anticancer drug discovery using natural compounds have been made in the last few decades, and polyphenols have emerged as promising molecules. They may act on various molecular targets because of their promiscuous behavior, presenting several physiological effects, some of which confer antitumor activity. This review analyzes the accumulated evidence of the antitumor effects of plant polyphenols on breast cancer, with special attention to their activity on ERs and HER2 targets and also covering different aspects such as redox balance, uncontrolled proliferation and chronic inflammation.
Collapse
Affiliation(s)
- María Losada-Echeberría
- Institute of Molecular and Cell Biology (IBMC), Miguel Hernández University (UMH), Avda. Universidad s/n, Elche 03202, Spain.
| | - María Herranz-López
- Institute of Molecular and Cell Biology (IBMC), Miguel Hernández University (UMH), Avda. Universidad s/n, Elche 03202, Spain.
| | - Vicente Micol
- Institute of Molecular and Cell Biology (IBMC), Miguel Hernández University (UMH), Avda. Universidad s/n, Elche 03202, Spain.
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), Palma de Mallorca 07122, Spain.
| | - Enrique Barrajón-Catalán
- Institute of Molecular and Cell Biology (IBMC), Miguel Hernández University (UMH), Avda. Universidad s/n, Elche 03202, Spain.
| |
Collapse
|
29
|
Hoang KV, Adcox HE, Fitch JR, Gordon DM, Curry HM, Schlesinger LS, White P, Gunn JS. AR-13, a Celecoxib Derivative, Directly Kills Francisella In Vitro and Aids Clearance and Mouse Survival In Vivo. Front Microbiol 2017; 8:1695. [PMID: 28955308 PMCID: PMC5600997 DOI: 10.3389/fmicb.2017.01695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/23/2017] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis (F. tularensis) is the causative agent of tularemia and is classified as a Tier 1 select agent. No licensed vaccine is currently available in the United States and treatment of tularemia is confined to few antibiotics. In this study, we demonstrate that AR-13, a derivative of the cyclooxygenase-2 inhibitor celecoxib, exhibits direct in vitro bactericidal killing activity against Francisella including a type A strain of F. tularensis (SchuS4) and the live vaccine strain (LVS), as well as toward the intracellular proliferation of LVS in macrophages, without causing significant host cell toxicity. Identification of an AR-13-resistant isolate indicates that this compound has an intracellular target(s) and that efflux pumps can mediate AR-13 resistance. In the mouse model of tularemia, AR-13 treatment protected 50% of the mice from lethal LVS infection and prolonged survival time from a lethal dose of F. tularensis SchuS4. Combination of AR-13 with a sub-optimal dose of gentamicin protected 60% of F. tularensis SchuS4-infected mice from death. Taken together, these data support the translational potential of AR-13 as a lead compound for the further development of new anti-Francisella agents.
Collapse
Affiliation(s)
- Ky V Hoang
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Haley E Adcox
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - James R Fitch
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States
| | - David M Gordon
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States
| | - Heather M Curry
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Peter White
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States.,Department of Pediatrics, The Ohio State University College of Medicine, ColumbusOH, United States
| | - John S Gunn
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| |
Collapse
|
30
|
The tumor suppressor LKB1 regulates starvation-induced autophagy under systemic metabolic stress. Sci Rep 2017; 7:7327. [PMID: 28779098 PMCID: PMC5544676 DOI: 10.1038/s41598-017-07116-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved process that degrades cellular components to restore energy homeostasis under limited nutrient conditions. How this starvation-induced autophagy is regulated at the whole-body level is not fully understood. Here, we show that the tumor suppressor Lkb1, which activates the key energy sensor AMPK, also regulates starvation-induced autophagy at the organismal level. Lkb1-deficient zebrafish larvae fail to activate autophagy in response to nutrient restriction upon yolk termination, shown by reduced levels of the autophagy-activating proteins Atg5, Lc3-II and Becn1, and aberrant accumulation of the cargo receptor and autophagy substrate p62. We demonstrate that the autophagy defect in lkb1 mutants can be partially rescued by inhibiting mTOR signaling but not by inhibiting the PI3K pathway. Interestingly, mTOR-independent activation of autophagy restores degradation of the aberrantly accumulated p62 in lkb1 mutants and prolongs their survival. Our data uncover a novel critical role for Lkb1 in regulating starvation-induced autophagy at the organismal level, providing mechanistic insight into metabolic adaptation during development.
Collapse
|
31
|
Munguia J, Nizet V. Pharmacological Targeting of the Host-Pathogen Interaction: Alternatives to Classical Antibiotics to Combat Drug-Resistant Superbugs. Trends Pharmacol Sci 2017; 38:473-488. [PMID: 28283200 DOI: 10.1016/j.tips.2017.02.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/17/2023]
Abstract
The rise of multidrug-resistant pathogens and the dearth of new antibiotic development place an existential strain on successful infectious disease therapy. Breakthrough strategies that go beyond classical antibiotic mechanisms are needed to combat this looming public health catastrophe. Reconceptualizing antibiotic therapy in the richer context of the host-pathogen interaction is required for innovative solutions. By defining specific virulence factors, the essence of a pathogen, and pharmacologically neutralizing their activities, one can block disease progression and sensitize microbes to immune clearance. Likewise, host-directed strategies to boost phagocyte bactericidal activity, enhance leukocyte recruitment, or reverse pathogen-induced immunosuppression seek to replicate the success of cancer immunotherapy in the field of infectious diseases. The answer to the threat of multidrug-resistant pathogens lies 'outside the box' of current antibiotic paradigms.
Collapse
Affiliation(s)
- Jason Munguia
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
32
|
Bachelder EM, Pino EN, Ainslie KM. Acetalated Dextran: A Tunable and Acid-Labile Biopolymer with Facile Synthesis and a Range of Applications. Chem Rev 2016; 117:1915-1926. [PMID: 28032507 DOI: 10.1021/acs.chemrev.6b00532] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acetalated dextran (Ac-DEX) is a tunable acid-labile biopolymer with facile synthesis, aptly designed for the formulation of microparticles for vaccines and immune modulation. Tunability of degradation is achieved based on the kinetics of reaction and the molecular weight of the parent dextran polymer. This tunability translated to differential rates of activation of CD8+ T cells in an in vitro ovalbumin model and illustrated that acid-labile polymer can activate CD8+ T cells at an increased rate compared to acid-insensitive polymers. In addition, Ac-DEX has been used to encapsulate small molecules, deliver nucleotides, transport inorganic molecules, formulate immune modulating therapies and vaccines, and trigger pH responsive constructs for therapy. Here we highlight the properties and results of Ac-DEX nano-/microparticles as well as the use of the polymer in other constructs and chemistries.
Collapse
Affiliation(s)
- Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Erica N Pino
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
33
|
Qi X, Man SM, Malireddi RKS, Karki R, Lupfer C, Gurung P, Neale G, Guy CS, Lamkanfi M, Kanneganti TD. Cathepsin B modulates lysosomal biogenesis and host defense against Francisella novicida infection. J Exp Med 2016; 213:2081-97. [PMID: 27551156 PMCID: PMC5030800 DOI: 10.1084/jem.20151938] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 07/22/2016] [Indexed: 12/11/2022] Open
Abstract
Lysosomal cathepsins regulate an exquisite range of biological functions, and their deregulation is associated with inflammatory, metabolic, and degenerative diseases in humans. In this study, we identified a key cell-intrinsic role for cathepsin B as a negative feedback regulator of lysosomal biogenesis and autophagy. Mice and macrophages lacking cathepsin B activity had increased resistance to the cytosolic bacterial pathogen Francisella novicida Genetic deletion or pharmacological inhibition of cathepsin B down-regulated mechanistic target of rapamycin activity and prevented cleavage of the lysosomal calcium channel TRPML1. These events drove transcription of lysosomal and autophagy genes via transcription factor EB, which increased lysosomal biogenesis and activation of autophagy initiation kinase ULK1 for clearance of the bacteria. Our results identified a fundamental biological function of cathepsin B in providing a checkpoint for homeostatic maintenance of lysosome populations and basic recycling functions in the cell.
Collapse
Affiliation(s)
- Xiaopeng Qi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Si Ming Man
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Christopher Lupfer
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Prajwal Gurung
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Mohamed Lamkanfi
- Inflammation Research Center, VIB, B-9052 Zwijnaarde-Ghent, Belgium Department of Internal Medicine, Ghent University, B-9000 Ghent, Belgium
| | | |
Collapse
|
34
|
Koselny K, Green J, Favazzo L, Glazier V, DiDone L, Ransford S, Krysan DJ. Antitumor/Antifungal Celecoxib Derivative AR-12 is a Non-Nucleoside Inhibitor of the ANL-Family Adenylating Enzyme Acetyl CoA Synthetase. ACS Infect Dis 2016; 2:268-280. [PMID: 27088128 PMCID: PMC4828684 DOI: 10.1021/acsinfecdis.5b00134] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Indexed: 02/08/2023]
Abstract
AR-12/OSU-03012 is an antitumor celecoxib-derivative that has progressed to Phase I clinical trial as an anticancer agent and has activity against a number of infectious agents including fungi, bacteria and viruses. However, the mechanism of these activities has remained unclear. Based on a chemical-genetic profiling approach in yeast, we have found that AR-12 is an ATP-competitive, time-dependent inhibitor of yeast acetyl coenzyme A synthetase. AR-12-treated fungal cells show phenotypes consistent with the genetic reduction of acetyl CoA synthetase activity, including induction of autophagy, decreased histone acetylation, and loss of cellular integrity. In addition, AR-12 is a weak inhibitor of human acetyl CoA synthetase ACCS2. Acetyl CoA synthetase activity is essential in many fungi and parasites. In contrast, acetyl CoA is primarily synthesized by an alternate enzyme, ATP-citrate lyase, in mammalian cells. Taken together, our results indicate that AR-12 is a non-nucleoside acetyl CoA synthetase inhibitor and that acetyl CoA synthetase may be a feasible antifungal drug target.
Collapse
Affiliation(s)
- Kristy Koselny
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Julianne Green
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Lacey Favazzo
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Virginia
E. Glazier
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Louis DiDone
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Shea Ransford
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Damian J. Krysan
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| |
Collapse
|
35
|
Needle-Free Delivery of Acetalated Dextran-Encapsulated AR-12 Protects Mice from Francisella tularensis Lethal Challenge. Antimicrob Agents Chemother 2016; 60:2052-62. [PMID: 26787696 DOI: 10.1128/aac.02228-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/06/2016] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensiscauses tularemia and is a potential biothreat. Given the limited antibiotics for treating tularemia and the possible use of antibiotic-resistant strains as a biowarfare agent, new antibacterial agents are needed. AR-12 is an FDA-approved investigational new drug (IND) compound that induces autophagy and has shown host-directed, broad-spectrum activityin vitroagainstSalmonella entericaserovar Typhimurium andF. tularensis We have shown that AR-12 encapsulated within acetalated dextran (Ace-DEX) microparticles (AR-12/MPs) significantly reduces host cell cytotoxicity compared to that with free AR-12, while retaining the ability to controlS.Typhimurium within infected human macrophages. In the present study, the toxicity and efficacy of AR-12/MPs in controlling virulent type AF. tularensisSchuS4 infection were examinedin vitroandin vivo No significant toxicity of blank MPs or AR-12/MPs was observed in lung histology sections when the formulations were given intranasally to uninfected mice. In histology sections from the lungs of intranasally infected mice treated with the formulations, increased macrophage infiltration was observed for AR-12/MPs, with or without suboptimal gentamicin treatment, but not for blank MPs, soluble AR-12, or suboptimal gentamicin alone. AR-12/MPs dramatically reduced the burden ofF. tularensisin infected human macrophages, in a manner similar to that of free AR-12. However,in vivo, AR-12/MPs significantly enhanced the survival ofF. tularensisSchuS4-infected mice compared to that seen with free AR-12. In combination with suboptimal gentamicin treatment, AR-12/MPs further improved the survival ofF. tularensisSchuS4-infected mice. These studies provide support for Ace-DEX-encapsulated AR-12 as a promising new therapeutic agent for tularemia.
Collapse
|
36
|
Law BYK, Mok SWF, Wu AG, Lam CWK, Yu MXY, Wong VKW. New Potential Pharmacological Functions of Chinese Herbal Medicines via Regulation of Autophagy. Molecules 2016; 21:359. [PMID: 26999089 PMCID: PMC6274228 DOI: 10.3390/molecules21030359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a universal catabolic cellular process for quality control of cytoplasm and maintenance of cellular homeostasis upon nutrient deprivation and environmental stimulus. It involves the lysosomal degradation of cellular components such as misfolded proteins or damaged organelles. Defects in autophagy are implicated in the pathogenesis of diseases including cancers, myopathy, neurodegenerations, infections and cardiovascular diseases. In the recent decade, traditional drugs with new clinical applications are not only commonly found in Western medicines, but also highlighted in Chinese herbal medicines (CHM). For instance, pharmacological studies have revealed that active components or fractions from Chaihu (Radix bupleuri), Hu Zhang (Rhizoma polygoni cuspidati), Donglingcao (Rabdosia rubesens), Hou po (Cortex magnoliae officinalis) and Chuan xiong (Rhizoma chuanxiong) modulate cancers, neurodegeneration and cardiovascular disease via autophagy. These findings shed light on the potential new applications and formulation of CHM decoctions via regulation of autophagy. This article reviews the roles of autophagy in the pharmacological actions of CHM and discusses their new potential clinical applications in various human diseases.
Collapse
Affiliation(s)
- Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Simon Wing Fai Mok
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - An Guo Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Christopher Wai Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Margaret Xin Yi Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
37
|
Collier MA, Peine KJ, Gautam S, Oghumu S, Varikuti S, Borteh H, Papenfuss TL, Sataoskar AR, Bachelder EM, Ainslie KM. Host-mediated Leishmania donovani treatment using AR-12 encapsulated in acetalated dextran microparticles. Int J Pharm 2016; 499:186-194. [PMID: 26768723 DOI: 10.1016/j.ijpharm.2016.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/15/2022]
Abstract
Leishmaniasis is a disease caused by parasites of Leishmania sp., which effects nearly 12 million people worldwide and is associated with treatment complications due to widespread parasite resistance toward pathogen-directed therapeutics. The current treatments for visceral leishmaniasis (VL), the systemic form of the disease, involve pathogen-mediated drugs and have long treatment regimens, increasing the risk of forming resistant strains. One way to limit emergence of resistant pathogens is through the use of host-mediated therapeutics. The host-mediated therapeutic AR-12, which is FDA IND-approved for cancer treatment, has shown activity against a broad spectrum of intracellular pathogens; however, due to hydrophobicity and toxicity, it is difficult to reach therapeutic doses. We have formulated AR-12 into microparticles (AR-12/MPs) using the novel biodegradable polymer acetalated dextran (Ace-DEX) and used this formulation for the systemic treatment of VL. Treatment with AR-12/MPs significantly reduced liver, spleen, and bone marrow parasite loads in infected mice, while combinatorial therapies with amphotericin B had an even more significant effect. Overall, AR-12/MPs offer a unique, host-mediated therapy that could significantly reduce the emergence of drug resistance in the treatment of VL.
Collapse
Affiliation(s)
- M A Collier
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - K J Peine
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - S Gautam
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - S Oghumu
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - S Varikuti
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - H Borteh
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - T L Papenfuss
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - A R Sataoskar
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - E M Bachelder
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - K M Ainslie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
38
|
Chang HC, Huang YT, Chen CS, Chen YW, Huang YT, Su JC, Teng LJ, Shiau CW, Chiu HC. In vitro and in vivo activity of a novel sorafenib derivative SC5005 against MRSA. J Antimicrob Chemother 2015; 71:449-59. [PMID: 26553845 DOI: 10.1093/jac/dkv367] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The emergence of MRSA strains resistant to most antibiotics is a serious threat to public health. Based on our discovery that the tyrosine kinase inhibitor sorafenib exhibits inhibitory activity against Staphylococcus species, the objective of this study is to exploit this unique antibacterial activity of sorafenib to develop novel antibacterial agents against MRSA. METHODS A sorafenib-based focused compound library was synthesized by substituting the pyridinyl and phenyl groups with different functional groups. The resulting sorafenib derivatives were screened for growth-suppressive activities against Staphylococcus aureus and Staphylococcus epidermidis following CLSI guidelines and for cytotoxicity towards human cells using MTT cell viability assays. Compounds with high selectivity for bacterial inhibition over cytotoxicity were further evaluated by time-kill assay and Caenorhabditis elegans and mice survival assays to evaluate their efficacy in vitro and in vivo. RESULTS The screening of sorafenib derivatives led to the identification of compound SC5005 as a lead compound with high potency in killing different clinical strains of MRSA with an MIC90 of 0.5 mg/L and with low cytotoxicity, as demonstrated by IC50-to-MIC ratios of up to 40. In addition, SC5005 showed a significant protective effect in MSSA- or MRSA-infected C. elegans. Intraperitoneal administration of SC5005 at 10 mg/kg significantly improved the survival of MRSA-infected C57BL/6 mice. CONCLUSIONS In light of its high potency in suppressing MRSA in both in vitro and in vivo models, SC5005 represents a potential lead agent for continued preclinical development as a therapeutic intervention against MRSA.
Collapse
Affiliation(s)
- Han-Chu Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chang-Shi Chen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Wei Chen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Tsung Huang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Jung-Chen Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Lee-Jeng Teng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Chieh Chiu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
39
|
Roberts JL, Tavallai M, Nourbakhsh A, Fidanza A, Cruz-Luna T, Smith E, Siembida P, Plamondon P, Cycon KA, Doern CD, Booth L, Dent P. GRP78/Dna K Is a Target for Nexavar/Stivarga/Votrient in the Treatment of Human Malignancies, Viral Infections and Bacterial Diseases. J Cell Physiol 2015; 230:2552-78. [PMID: 25858032 PMCID: PMC4843173 DOI: 10.1002/jcp.25014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 01/10/2023]
Abstract
Prior tumor cell studies have shown that the drugs sorafenib (Nexavar) and regorafenib (Stivarga) reduce expression of the chaperone GRP78. Sorafenib/regorafenib and the multi‐kinase inhibitor pazopanib (Votrient) interacted with sildenafil (Viagra) to further rapidly reduce GRP78 levels in eukaryotes and as single agents to reduce Dna K levels in prokaryotes. Similar data were obtained in tumor cells in vitro and in drug‐treated mice for: HSP70, mitochondrial HSP70, HSP60, HSP56, HSP40, HSP10, and cyclophilin A. Prolonged ‘rafenib/sildenafil treatment killed tumor cells and also rapidly decreased the expression of: the drug efflux pumps ABCB1 and ABCG2; and NPC1 and NTCP, receptors for Ebola/Hepatitis A and B viruses, respectively. Pre‐treatment with the ‘Rafenib/sildenafil combination reduced expression of the Coxsackie and Adenovirus receptor in parallel with it also reducing the ability of a serotype 5 Adenovirus or Coxsackie virus B4 to infect and to reproduce. Sorafenib/pazopanib and sildenafil was much more potent than sorafenib/pazopanib as single agents at preventing Adenovirus, Mumps, Chikungunya, Dengue, Rabies, West Nile, Yellow Fever, and Enterovirus 71 infection and reproduction. ‘Rafenib drugs/pazopanib as single agents killed laboratory generated antibiotic resistant E. coli which was associated with reduced Dna K and Rec A expression. Marginally toxic doses of ‘Rafenib drugs/pazopanib restored antibiotic sensitivity in pan‐antibiotic resistant bacteria including multiple strains of blakpcKlebsiella pneumoniae. Thus, Dna K is an antibiotic target for sorafenib, and inhibition of GRP78/Dna K has therapeutic utility for cancer and for bacterial and viral infections. J. Cell. Physiol. 230: 2552–2578, 2015. © 2015 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Aida Nourbakhsh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | - Christopher D Doern
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
40
|
Cano V, March C, Insua JL, Aguiló N, Llobet E, Moranta D, Regueiro V, Brennan GP, Millán-Lou MI, Martín C, Garmendia J, Bengoechea JA. Klebsiella pneumoniaesurvives within macrophages by avoiding delivery to lysosomes. Cell Microbiol 2015; 17:1537-60. [DOI: 10.1111/cmi.12466] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/22/2015] [Accepted: 05/28/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Victoria Cano
- Laboratory Infection and Immunity; Fundació d'Investigació Sanitària de les Illes Balears (FISIB); Bunyola Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
| | - Catalina March
- Laboratory Infection and Immunity; Fundació d'Investigació Sanitària de les Illes Balears (FISIB); Bunyola Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
| | - Jose Luis Insua
- Centre for Infection and Immunity; Queen's University Belfast; Belfast UK
| | - Nacho Aguiló
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública; Universidad de Zaragoza; Zaragoza Spain
| | - Enrique Llobet
- Laboratory Infection and Immunity; Fundació d'Investigació Sanitària de les Illes Balears (FISIB); Bunyola Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Institut d'Investigació Sanitària de Palma (IdISPa); Palma Spain
| | - David Moranta
- Laboratory Infection and Immunity; Fundació d'Investigació Sanitària de les Illes Balears (FISIB); Bunyola Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Institut d'Investigació Sanitària de Palma (IdISPa); Palma Spain
| | - Verónica Regueiro
- Laboratory Infection and Immunity; Fundació d'Investigació Sanitària de les Illes Balears (FISIB); Bunyola Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Institut d'Investigació Sanitària de Palma (IdISPa); Palma Spain
| | - Gerard P. Brennan
- School of Biological Sciences; Queen's University Belfast; Belfast UK
| | - Maria Isabel Millán-Lou
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública; Universidad de Zaragoza; Zaragoza Spain
| | - Carlos Martín
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública; Universidad de Zaragoza; Zaragoza Spain
| | - Junkal Garmendia
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES); Bunyola Spain
- Instituto de Agrobiotecnología; CSIC - Universidad Pública de Navarra-Gobierno de Navarra; Mutilva Spain
| | - José A. Bengoechea
- Centre for Infection and Immunity; Queen's University Belfast; Belfast UK
- Consejo Superior de Investigaciones Científicas (CSIC); Madrid Spain
| |
Collapse
|
41
|
Booth L, Roberts JL, Cash DR, Tavallai S, Jean S, Fidanza A, Cruz-Luna T, Siembiba P, Cycon KA, Cornelissen CN, Dent P. GRP78/BiP/HSPA5/Dna K is a universal therapeutic target for human disease. J Cell Physiol 2015; 230:1661-76. [PMID: 25546329 PMCID: PMC4402027 DOI: 10.1002/jcp.24919] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 01/11/2023]
Abstract
The chaperone GRP78/Dna K is conserved throughout evolution down to prokaryotes. The GRP78 inhibitor OSU-03012 (AR-12) interacted with sildenafil (Viagra) or tadalafil (Cialis) to rapidly reduce GRP78 levels in eukaryotes and as a single agent reduce Dna K levels in prokaryotes. Similar data with the drug combination were obtained for: HSP70, HSP90, GRP94, GRP58, HSP27, HSP40 and HSP60. OSU-03012/sildenafil treatment killed brain cancer stem cells and decreased the expression of: NPC1 and TIM1; LAMP1; and NTCP1, receptors for Ebola/Marburg/Hepatitis A, Lassa fever, and Hepatitis B viruses, respectively. Pre-treatment with OSU-03012/sildenafil reduced expression of the coxsakie and adenovirus receptor in parallel with it also reducing the ability of a serotype 5 adenovirus or coxsakie virus B4 to infect and to reproduce. Similar data were obtained using Chikungunya, Mumps, Measles, Rubella, RSV, CMV, and Influenza viruses. OSU-03012 as a single agent at clinically relevant concentrations killed laboratory generated antibiotic resistant E. coli and clinical isolate multi-drug resistant N. gonorrhoeae and MRSE which was in bacteria associated with reduced Dna K and Rec A expression. The PDE5 inhibitors sildenafil or tadalafil enhanced OSU-03012 killing in N. gonorrhoeae and MRSE and low marginally toxic doses of OSU-03012 could restore bacterial sensitivity in N. gonorrhoeae to multiple antibiotics. Thus, Dna K and bacterial phosphodiesterases are novel antibiotic targets, and inhibition of GRP78 is of therapeutic utility for cancer and also for bacterial and viral infections. J. Cell. Physiol. 230: 1661–1676, 2015. © 2014 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yu JY, Zhang B, Peng L, Wu CH, Cao H, Zhong JF, Hoffman J, Huang SH. Repositioning of Memantine as a Potential Novel Therapeutic Agent against Meningitic E. coli-Induced Pathogenicities through Disease-Associated Alpha7 Cholinergic Pathway and RNA Sequencing-Based Transcriptome Analysis of Host Inflammatory Responses. PLoS One 2015; 10:e0121911. [PMID: 25993608 PMCID: PMC4437645 DOI: 10.1371/journal.pone.0121911] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/07/2015] [Indexed: 01/09/2023] Open
Abstract
Neonatal sepsis and meningitis (NSM) remains a leading cause worldwide of mortality and morbidity in newborn infants despite the availability of antibiotics over the last several decades. E. coli is the most common gram-negative pathogen causing NSM. Our previous studies show that α7 nicotinic receptor (α7 nAChR), an essential regulator of inflammation, plays a detrimental role in the host defense against NSM. Despite notable successes, there still exists an unmet need for new effective therapeutic approaches to treat this disease. Using the in vitro/in vivo models of the blood-brain barrier (BBB) and RNA-seq, we undertook a drug repositioning study to identify unknown antimicrobial activities for known drugs. We have demonstrated for the first time that memantine (MEM), a FDA-approved drug for treatment of Alzheimer’s disease, could very efficiently block E. coli-caused bacteremia and meningitis in a mouse model of NSM in a manner dependent on α7 nAChR. MEM was able to synergistically enhance the antibacterial activity of ampicillin in HBMEC infected with E. coli K1 (E44) and in neonatal mice with E44-caused bacteremia and meningitis. Differential gene expression analysis of RNA-Seq data from mouse BMEC infected with E. coli K1 showed that several E44-increased inflammatory factors, including IL33, IL18rap, MMP10 and Irs1, were significantly reduced by MEM compared to the infected cells without drug treatment. MEM could also significantly up-regulate anti-inflammatory factors, including Tnfaip3, CISH, Ptgds and Zfp36. Most interestingly, these factors may positively and negatively contribute to regulation of NF-κB, which is a hallmark feature of bacterial meningitis. Furthermore, we have demonstrated that circulating BMEC (cBMEC) are the potential novel biomarkers for NSM. MEM could significantly reduce E44-increased blood level of cBMEC in mice. Taken together, our data suggest that memantine can efficiently block host inflammatory responses to bacterial infection through modulation of both inflammatory and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Jing-Yi Yu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Bao Zhang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Liang Peng
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America; Department of Clinic Laboratory, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Chun-Hua Wu
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - John F Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America; Department of Perio, Diagnostic Sciences & Biomedical Sciences, School of Dentistry, University of Southern California, Los Angeles, CA, 93003, United States of America; Department of Pediatrics, School of Medicine, University of Southern California, Los Angeles, CA, 93003, United States of America
| | - Jill Hoffman
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| |
Collapse
|
43
|
Booth L, Roberts JL, Dent P. HSPA5/Dna K may be a useful target for human disease therapies. DNA Cell Biol 2015; 34:153-8. [PMID: 25689303 DOI: 10.1089/dna.2015.2808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The chaperone protein HSPA5/Dna K is conserved throughout evolution from higher eukaryotes down to prokaryotes. The celecoxib derivative OSU-03012 (also called AR-12) interacts with Viagra or Cialis in eukaryotic cells to rapidly reduce HSPA5 levels as well as blunt the functions of many other chaperone proteins. Because multiple chaperones are modulated in eukaryotes, the expression of cell surface virus receptors is reduced and because HSPA5 in blocked viruses cannot efficiently replicate. Because DnaK levels are reduced in prokaryotes by OSU-03012, the levels of DnaK chaperone proteins such as Rec A decline, which is associated with bacterial cell death and a resensitization of so-called drug-resistant superbugs to standard of care antibiotics. In Alzheimer's disease, HSPA5 has been shown to play a supportive role for the progression of tau phosphorylation and neurodegeneration. Thus, in eukaryotes, HSPA5 represents a target for anticancer, antiviral, and anti-Alzheimer's therapeutics and in prokaryotes, DnaK and bacterial phosphodiesterases represent novel antibiotic targets that should be exploited in the future by pharmaceutical companies.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, Virginia
| | | | | |
Collapse
|
44
|
Salunke SB, Azad AK, Kapuriya NP, Balada-Llasat JM, Pancholi P, Schlesinger LS, Chen CS. Design and synthesis of novel anti-tuberculosis agents from the celecoxib pharmacophore. Bioorg Med Chem 2015; 23:1935-43. [PMID: 25818768 DOI: 10.1016/j.bmc.2015.03.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/03/2015] [Accepted: 03/15/2015] [Indexed: 11/15/2022]
Abstract
The identification of compounds with anti-mycobacterial activity within classes of molecules that have been developed for other purposes is a fruitful approach for the development of anti-tuberculosis (TB) agents. In this study we used the scaffold of celecoxib which exhibits several activities against different pathogens, for the design and focused synthesis of a library of 64 compounds. For the primary screen, we used a bioluminescence-based method by constructing a luciferase-expressing reporter M.tb strain which contains the entire bacterial Lux operon cloned in a mycobacterial integrative expression vector. Through the screening of this library, we identified 6 hit compounds with high in vitro anti-mycobacterial activity (IC₅₀ ∼0.18-0.48 μM). In particular, compounds 41, 51 and 53 were capable of inhibiting M.tb as effectively as the anti-TB drug isoniazid (INH) at 5 μM over a 72-h period, as analyzed by both bioluminescence- and colony forming unit (CFU)-based assays. All hit compounds also showed anti-M.tb activities against several multi-drug-resistant (MDR) strains. Most of the hit compounds showed no cytotoxicity for human macrophages at concentrations as high as 40 μM, setting the stage for further optimization and development of these anti-TB hit compounds both ex vivo and in vivo.
Collapse
Affiliation(s)
- Santosh B Salunke
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States
| | - Abul K Azad
- Center for Microbial Interface Biology, The Ohio State University, 460 W 12th Avenue, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Wexner Medical Center, 460 W 12th Avenue, Columbus, OH 43210, United States
| | - Naval P Kapuriya
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States; Division of Pharmaceutical and Organic Chemistry, M. & N. Virani Science College, Saurashtra University, Rajkot 360005, Gujarat, India
| | - Joan-Miquel Balada-Llasat
- Department of Pathology, The Ohio State University, 410 W. 10th Avenue, Columbus, OH 43210, United States
| | - Preeti Pancholi
- Department of Pathology, The Ohio State University, 410 W. 10th Avenue, Columbus, OH 43210, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, The Ohio State University, 460 W 12th Avenue, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Wexner Medical Center, 460 W 12th Avenue, Columbus, OH 43210, United States.
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, United States; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
45
|
Hoang KV, Borteh HM, Rajaram MVS, Peine KJ, Curry H, Collier MA, Homsy ML, Bachelder EM, Gunn JS, Schlesinger LS, Ainslie KM. Acetalated dextran encapsulated AR-12 as a host-directed therapy to control Salmonella infection. Int J Pharm 2014; 477:334-43. [PMID: 25447826 DOI: 10.1016/j.ijpharm.2014.10.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/22/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
AR-12 has been evaluated in clinical trials as an anti-cancer agent but also has demonstrated host-directed, broad-spectrum clearance of bacteria. We have previously shown that AR-12 has activity in vitro against Salmonella enterica serovar Typhimurium and Francisella species by inducing autophagy and other host immune pathways. AR-12 treatment of S. Typhimurium-infected mice resulted in a 10-fold reduction in bacterial load in the liver and spleen and an increased survival time. However, AR-12 treatment did not protect mice from death, likely due poor formulation. In the current study, AR-12 was encapsulated in a microparticulate carrier formulated from the novel degradable biopolymer acetalated dextran (Ace-DEX) and subsequently evaluated for its activity in human monocyte-derived macrophages (hMDMs). Our results show that hMDMs efficiently internalized Ace-DEX microparticles (MPs), and that encapsulation significantly reduced host cell cytotoxicity compared to unencapsulated AR-12. Efficient macrophage internalization of AR-12 loaded MPs (AR-12/MPs) was further demonstrated by autophagosome formation that was comparable to free AR-12 and resulted in enhanced clearance of intracellular Salmonella. Taken together, these studies provide support that Ace-DEX encapsulated AR-12 may be a promising new therapeutic agent to control intracellular bacterial pathogens of macrophages by targeting delivery and reducing drug toxicity.
Collapse
Affiliation(s)
- Ky V Hoang
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| | - Hassan M Borteh
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| | - Kevin J Peine
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Heather Curry
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| | - Michael A Collier
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Michael L Homsy
- Department of Chemical and Bimolecular Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Eric M Bachelder
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - John S Gunn
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| | - Kristy M Ainslie
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC 27599, United States.
| |
Collapse
|
46
|
Sensitization of intracellular Salmonella enterica serovar Typhimurium to aminoglycosides in vitro and in vivo by a host-targeted antimicrobial agent. Antimicrob Agents Chemother 2014; 58:7375-82. [PMID: 25267669 DOI: 10.1128/aac.03778-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Aminoglycosides exhibit relatively poor activity against intracellular Salmonella enterica serovar Typhimurium due to their low permeativity across eukaryotic cell membranes. Previously, we identified the unique ability of AR-12, a celecoxib-derived small-molecule agent, to eradicate intracellular Salmonella Typhimurium in macrophages by facilitating autophagosome formation and suppressing Akt kinase signaling. In light of this unique mode of antibacterial action, we investigated the ability of AR-12 to sensitize intracellular Salmonella to aminoglycosides in macrophages and in an animal model. The antibacterial activities of AR-12 combined with various aminoglycosides, including streptomycin, kanamycin, gentamicin, and amikacin, against intracellular S. Typhimurium in murine RAW264.7 macrophages were assessed. Cells were infected with S. Typhimurium followed by treatment with AR-12 or individual aminoglycosides or with combinations for 24 h. The in vivo efficacies of AR-12, alone or in combination with gentamicin or amikacin, were also assessed by treating S. Typhimurium-infected BALB/c mice daily for 14 consecutive days. Exposure of S. Typhimurium-infected RAW264.7 cells to a combination of AR-12 with individual aminoglycosides led to a reduction in bacterial survival (P < 0.05), both intracellular and extracellular, that was greater than that seen with the aminoglycosides alone. This sensitizing effect, however, was not associated with increased aminoglycoside penetration into bacteria or macrophages. Moreover, daily intraperitoneal injection of AR-12 at 0.1 mg/kg of body weight significantly increased the in vivo efficacy of gentamicin and amikacin in prolonging the survival of S. Typhimurium-infected mice. These findings indicate that the unique ability of AR-12 to enhance the in vivo efficacy of aminoglycosides might have translational potential for efforts to develop novel strategies for the treatment of salmonellosis.
Collapse
|
47
|
The impact of "omic" and imaging technologies on assessing the host immune response to biodefence agents. J Immunol Res 2014; 2014:237043. [PMID: 25333059 PMCID: PMC4182007 DOI: 10.1155/2014/237043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023] Open
Abstract
Understanding the interactions between host and pathogen is important for the development and assessment of medical countermeasures to infectious agents, including potential biodefence pathogens such as Bacillus anthracis, Ebola virus, and Francisella tularensis. This review focuses on technological advances which allow this interaction to be studied in much greater detail. Namely, the use of “omic” technologies (next generation sequencing, DNA, and protein microarrays) for dissecting the underlying host response to infection at the molecular level; optical imaging techniques (flow cytometry and fluorescence microscopy) for assessing cellular responses to infection; and biophotonic imaging for visualising the infectious disease process. All of these technologies hold great promise for important breakthroughs in the rational development of vaccines and therapeutics for biodefence agents.
Collapse
|
48
|
van der Vaart M, Korbee C, Lamers G, Tengeler A, Hosseini R, Haks M, Ottenhoff T, Spaink H, Meijer A. The DNA Damage-Regulated Autophagy Modulator DRAM1 Links Mycobacterial Recognition via TLR-MYD88 to Autophagic Defense. Cell Host Microbe 2014; 15:753-67. [DOI: 10.1016/j.chom.2014.05.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 01/28/2014] [Accepted: 04/24/2014] [Indexed: 02/02/2023]
|
49
|
Härtlova A, Link M, Balounova J, Benesova M, Resch U, Straskova A, Sobol M, Philimonenko A, Hozak P, Krocova Z, Gekara N, Filipp D, Stulik J. Quantitative proteomics analysis of macrophage-derived lipid rafts reveals induction of autophagy pathway at the early time of Francisella tularensis LVS infection. J Proteome Res 2014; 13:796-804. [PMID: 24364512 DOI: 10.1021/pr4008656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Francisella tularensis is a highly infectious intracellular pathogen that has evolved an efficient strategy to subvert host defense response to survive inside the host. The molecular mechanisms regulating these host-pathogen interactions and especially those that are initiated at the time of the bacterial entry via its attachment to the host plasma membrane likely predetermine the intracellular fate of pathogen. Here, we provide the evidence that infection of macrophages with F. tularensis leads to changes in protein composition of macrophage-derived lipid rafts, isolated as detergent-resistant membranes (DRMs). Using SILAC-based quantitative proteomic approach, we observed the accumulation of autophagic adaptor protein p62 at the early stages of microbe-host cell interaction. We confirmed the colocalization of the p62 with ubiquitinated and LC3-decorated intracellular F. tularensis microbes with its maximum at 1 h postinfection. Furthermore, the infection of p62-knockdown host cells led to the transient increase in the intracellular number of microbes up to 4 h after in vitro infection. Together, these data suggest that the activation of the autophagy pathway in F. tularensis infected macrophages, which impacts the early phase of microbial proliferation, is subsequently circumvented by ongoing infection.
Collapse
Affiliation(s)
- Anetta Härtlova
- Centre of Advanced Studies , Faculty of Military Health Sciences, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Al-Khodor S, Marshall-Batty K, Nair V, Ding L, Greenberg DE, Fraser IDC. Burkholderia cenocepacia J2315 escapes to the cytosol and actively subverts autophagy in human macrophages. Cell Microbiol 2013; 16:378-95. [PMID: 24119232 DOI: 10.1111/cmi.12223] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/27/2013] [Accepted: 10/04/2013] [Indexed: 12/15/2022]
Abstract
Selective autophagy functions to specifically degrade cellular cargo tagged by ubiquitination, including bacteria. Strains of the Burkholderia cepacia complex (Bcc) are opportunistic pathogens that cause life-threatening infections in patients with cystic fibrosis (CF) and chronic granulomatous disease (CGD). While there is evidence that defective macrophage autophagy in a mouse model of CF can influence B. cenocepacia susceptibility, there have been no comprehensive studies on how this bacterium is sensed and targeted by the host autophagy response in human macrophages. Here, we describe the intracellular life cycle of B. cenocepacia J2315 and its interaction with the autophagy pathway in human cells. Electron and confocal microscopy analyses demonstrate that the invading bacteria interact transiently with the endocytic pathway before escaping to the cytosol. This escape triggers theselective autophagy pathway, and the recruitment of ubiquitin, the ubiquitin-binding adaptors p62 and NDP52 and the autophagosome membrane-associated protein LC3B, to the bacterial vicinity. However, despite recruitment of these key autophagy pathway effectors, B. cenocepacia blocks autophagosome completion and replicates in the host cytosol. We find that a pre-infection increase in cellular autophagy flux can significantly inhibit B. cenocepacia replication and that lower autophagy flux in macrophages from immunocompromised CGD patients could contribute to increased B. cenocepacia susceptibility, identifying autophagy manipulation as a potential therapeutic approach to reduce bacterial burden in B. cenocepacia infections.
Collapse
Affiliation(s)
- Souhaila Al-Khodor
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | | | | | |
Collapse
|