51
|
Asdaq SMB, Ikbal AMA, Sahu RK, Bhattacharjee B, Paul T, Deka B, Fattepur S, Widyowati R, Vijaya J, Al mohaini M, Alsalman AJ, Imran M, Nagaraja S, Nair AB, Attimarad M, Venugopala KN. Nanotechnology Integration for SARS-CoV-2 Diagnosis and Treatment: An Approach to Preventing Pandemic. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1841. [PMID: 34361227 PMCID: PMC8308419 DOI: 10.3390/nano11071841] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
The SARS-CoV-2 outbreak is the COVID-19 disease, which has caused massive health devastation, prompting the World Health Organization to declare a worldwide health emergency. The corona virus infected millions of people worldwide, and many died as a result of a lack of particular medications. The current emergency necessitates extensive therapy in order to stop the spread of the coronavirus. There are various vaccinations available, but no validated COVID-19 treatments. Since its outbreak, many therapeutics have been tested, including the use of repurposed medications, nucleoside inhibitors, protease inhibitors, broad spectrum antivirals, convalescence plasma therapies, immune-modulators, and monoclonal antibodies. However, these approaches have not yielded any outcomes and are mostly used to alleviate symptoms associated with potentially fatal adverse drug reactions. Nanoparticles, on the other hand, may prove to be an effective treatment for COVID-19. They can be designed to boost the efficacy of currently available antiviral medications or to trigger a rapid immune response against COVID-19. In the last decade, there has been significant progress in nanotechnology. This review focuses on the virus's basic structure, pathogenesis, and current treatment options for COVID-19. This study addresses nanotechnology and its applications in diagnosis, prevention, treatment, and targeted vaccine delivery, laying the groundwork for a successful pandemic fight.
Collapse
Affiliation(s)
| | - Abu Md Ashif Ikbal
- Department of Pharmacy, Tripura University (A Central University), Suryamaninagar 799022, Tripura (W), India;
| | - Ram Kumar Sahu
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia;
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar 788011, Assam, India
| | - Bedanta Bhattacharjee
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (B.B.); (T.P.); (B.D.)
| | - Tirna Paul
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (B.B.); (T.P.); (B.D.)
| | - Bhargab Deka
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (B.B.); (T.P.); (B.D.)
| | - Santosh Fattepur
- School of Pharmacy, Management and Science University, Seksyen 13, Shah Alam 40100, Selangor, Malaysia
| | - Retno Widyowati
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia;
| | - Joshi Vijaya
- Department of Pharmaceutics, Government College of Pharmacy, Bangalore 560027, Karnataka, India;
| | - Mohammed Al mohaini
- Basic Sciences Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Alahsa 31982, Saudi Arabia;
- King Abdullah International Medical Research Center, Alahsa 31982, Saudi Arabia
| | - Abdulkhaliq J. Alsalman
- Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | - Sreeharsha Nagaraja
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia; (S.N.); (A.B.N.); (M.A.); (K.N.V.)
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bangalore 560035, India
| | - Anroop B. Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia; (S.N.); (A.B.N.); (M.A.); (K.N.V.)
| | - Mahesh Attimarad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia; (S.N.); (A.B.N.); (M.A.); (K.N.V.)
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia; (S.N.); (A.B.N.); (M.A.); (K.N.V.)
- Department of Biotechnology and Food Technology, Durban University of Technology, Durban 4001, South Africa
| |
Collapse
|
52
|
Yin H, Zhou M, Chen X, Wan TF, Jin L, Rao SS, Tan YJ, Duan R, Zhang Y, Wang ZX, Wang YY, He ZH, Luo MJ, Hu XK, Wang Y, Situ WY, Tang SY, Liu WE, Chen CY, Xie H. Fructose-coated Ångstrom silver prevents sepsis by killing bacteria and attenuating bacterial toxin-induced injuries. Am J Cancer Res 2021; 11:8152-8171. [PMID: 34373734 PMCID: PMC8344005 DOI: 10.7150/thno.55334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/28/2021] [Indexed: 11/29/2022] Open
Abstract
Serious infection caused by multi-drug-resistant bacteria is a major threat to human health. Bacteria can invade the host tissue and produce various toxins to damage or kill host cells, which may induce life-threatening sepsis. Here, we aimed to explore whether fructose-coated Ångstrom-scale silver particles (F-AgÅPs), which were prepared by our self-developed evaporation-condensation system and optimized coating approach, could kill bacteria and sequester bacterial toxins to attenuate fatal bacterial infections. Methods: A series of in vitro assays were conducted to test the anti-bacterial efficacy of F-AgÅPs, and to investigate whether F-AgÅPs could protect against multi-drug resistant Staphylococcus aureus (S. aureus)- and Escherichia coli (E. coli)-induced cell death, and suppress their toxins (S. aureus hemolysin and E. coli lipopolysaccharide)-induced cell injury or inflammation. The mouse models of cecal ligation and puncture (CLP)- or E. coli bloodstream infection-induced lethal sepsis were established to assess whether the intravenous administration of F-AgÅPs could decrease bacterial burden, inhibit inflammation, and improve the survival rates of mice. The levels of silver in urine and feces of mice were examined to evaluate the excretion of F-AgÅPs. Results: F-AgÅPs efficiently killed various bacteria that can cause lethal infections and also competed with host cells to bind with S. aureus α-hemolysin, thus blocking its cytotoxic activity. F-AgÅPs inhibited E. coli lipopolysaccharide-induced endothelial injury and macrophage inflammation, but not by directly binding to lipopolysaccharide. F-AgÅPs potently reduced bacterial burden, reversed dysregulated inflammation, and enhanced survival in mice with CLP- or E. coli bloodstream infection-induced sepsis, either alone or combined with antibiotic therapy. After three times injections within 48 h, 79.18% of F-AgÅPs were excreted via feces at the end of the 14-day observation period. Conclusion: This study suggests the prospect of F-AgÅPs as a promising intravenous agent for treating severe bacterial infections.
Collapse
|
53
|
Zhao Y, Pu M, Zhang J, Wang Y, Yan X, Yu L, He Z. Recent advancements of nanomaterial-based therapeutic strategies toward sepsis: bacterial eradication, anti-inflammation, and immunomodulation. NANOSCALE 2021; 13:10726-10747. [PMID: 34165483 DOI: 10.1039/d1nr02706a] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sepsis is a life threatening disease that is caused by a dysregulated host immune response to infection, resulting in tissue damage and organ dysfunction, which account for a high in-hospital mortality (approximately 20%). However, there are still no effective and specific therapeutics for clinical sepsis management. Nanomaterial-based strategies have emerged as promising tools for improving the therapeutic efficacy of sepsis by combating lethal bacterial infection, modulating systemic inflammatory response, preventing multiple organ failure, etc. This review has comprehensively summarized the recent advancements in nanomaterial-based strategies for the management of sepsis and severe complications, in which those nanosystems act either as inherent therapeutics or as nanocarriers for the precise delivery of agents. These formulations mechanically possess antibacterial, anti-inflammatory, immunomodulatory, and anti-oxidative effects, achieving multifunctional synergistic treatment efficacy against sepsis. Furthermore, several cell membrane-derived biomimetic nanoplatforms have been used as decoys to trap and neutralize the pathogenic toxins. The critical role of other adjuvant therapies in sepsis management, including the combination of nanotechnology and stem cell therapy, is also highlighted. Overall, this review provides insights into innovative nanotechnology-based strategies applied in sepsis treatment.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Minju Pu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Jingwen Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Yanan Wang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Xuefeng Yan
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Zhiyu He
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| |
Collapse
|
54
|
Tian M, Xing R, Guan J, Yang B, Zhao X, Yang J, Zhan C, Zhang S. A Nanoantidote Alleviates Glioblastoma Chemotoxicity without Efficacy Compromise. NANO LETTERS 2021; 21:5158-5166. [PMID: 34097422 DOI: 10.1021/acs.nanolett.1c01201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer patients suffer from the toxicity of chemotherapy. Antidote, given as a remedy limiting poison, is an effective way to counteract toxicity. However, few antidotes abrogate chemotoxicity without compromising the therapeutic efficacy. Herein, a rationally designed nanoantidote can neutralize chemo-agents in normal cells but not enter tumors and thus would not interfere with the efficacy of tumor treatment. The nanoantidote, consisting of a dendrimer core wrapped by reductive cysteine, captures Temozolomide (TMZ, the glioblastoma standard chemotherapy). Meanwhile, thanks to the blood-brain barrier (BBB) and the size of the nanoantidote, the nanoantidote cannot enter glioblastoma. In murine models, the nanoantidote distributes in normal tissues without crossing the BBB, so it markedly reduces the chemotoxicity of TMZ and retains the original TMZ therapeutic efficacy. With most nanotechnologies focusing on antitumor treatment, this detoxicating strategy demonstrates a nanoplatform to reduce chemotoxicity using physiology barriers and introduces a new approach to nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Meng Tian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Rui Xing
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Juan Guan
- Department of Pharmacology, School of Basic Medical Sciences and Center of Medical Research and Innovation, Shanghai Pudong Hospital and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P.R. China
| | - Bingxue Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Xin Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Juanjuan Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences and Center of Medical Research and Innovation, Shanghai Pudong Hospital and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P.R. China
| | - Shiyi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
55
|
Liposomes Prevent In Vitro Hemolysis Induced by Streptolysin O and Lysenin. MEMBRANES 2021; 11:membranes11050364. [PMID: 34069894 PMCID: PMC8157566 DOI: 10.3390/membranes11050364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022]
Abstract
The need for alternatives to antibiotics in the fight against infectious diseases has inspired scientists to focus on antivirulence factors instead of the microorganisms themselves. In this respect, prior work indicates that tiny, enclosed bilayer lipid membranes (liposomes) have the potential to compete with cellular targets for toxin binding, hence preventing their biological attack and aiding with their clearance. The effectiveness of liposomes as decoy targets depends on their availability in the host and how rapidly they are cleared from the circulation. Although liposome PEGylation may improve their circulation time, little is known about how such a modification influences their interactions with antivirulence factors. To fill this gap in knowledge, we investigated regular and long-circulating liposomes for their ability to prevent in vitro red blood cell hemolysis induced by two potent lytic toxins, lysenin and streptolysin O. Our explorations indicate that both regular and long-circulating liposomes are capable of similarly preventing lysis induced by streptolysin O. In contrast, PEGylation reduced the effectiveness against lysenin-induced hemolysis and altered binding dynamics. These results suggest that toxin removal by long-circulating liposomes is feasible, yet dependent on the particular virulence factor under scrutiny.
Collapse
|
56
|
Gkartziou F, Giormezis N, Spiliopoulou I, Antimisiaris SG. Nanobiosystems for Antimicrobial Drug-Resistant Infections. NANOMATERIALS 2021; 11:nano11051075. [PMID: 33922004 PMCID: PMC8143556 DOI: 10.3390/nano11051075] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 02/07/2023]
Abstract
The worldwide increased bacterial resistance toward antimicrobial therapeutics has led investigators to search for new therapeutic options. Some of the options currently exploited to treat drug-resistant infections include drug-associated nanosystems. Additionally, the use of bacteriophages alone or in combination with drugs has been recently revisited; some studies utilizing nanosystems for bacteriophage delivery have been already reported. In this review article, we focus on nine pathogens that are the leading antimicrobial drug-resistant organisms, causing difficult-to-treat infections. For each organism, the bacteriophages and nanosystems developed or used in the last 20 years as potential treatments of pathogen-related infections are discussed. Summarizing conclusions and future perspectives related with the potential of such nano-antimicrobials for the treatment of persistent infections are finally highlighted.
Collapse
Affiliation(s)
- Foteini Gkartziou
- Institute of Chemical Engineering, FORTH/ICES, Platani, 26504 Patras, Greece;
| | - Nikolaos Giormezis
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Iris Spiliopoulou
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
- Correspondence: (I.S.); (S.G.A.)
| | - Sophia G. Antimisiaris
- Institute of Chemical Engineering, FORTH/ICES, Platani, 26504 Patras, Greece;
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
- Correspondence: (I.S.); (S.G.A.)
| |
Collapse
|
57
|
Jiang L, Zhu Y, Luan P, Xu J, Ru G, Fu JG, Sang N, Xiong Y, He Y, Lin GQ, Wang J, Zhang J, Li R. Bacteria -Anchoring Hybrid Liposome Capable of Absorbing Multiple Toxins for Antivirulence Therapy of Escherichia coli Infection. ACS NANO 2021; 15:4173-4185. [PMID: 33606516 DOI: 10.1021/acsnano.0c04800] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antivirulence therapy by cell membrane coated nanoparticles has shown promise against bacterial infections. However, current approaches remain unsatisfactory when facing Escherichia coli (E. coli) infections, since the E. coli secretes multiple bacterial toxins including endotoxins and exotoxins that are challenging to eliminate simultaneously. What is worse, the absorptive scavengers normally rely on random contact of the diffuse toxins, which is not efficient. For the current cell membrane coated platform, the single type of cell membrane cannot fully meet the detoxing requirement facing multiple toxins. To address these problems, a polymyxin B (PMB)-modified, red blood cell (RBC)-mimetic hybrid liposome (P-RL) was developed. The P-RL was fabricated succinctly through fusion of PMB-modified lipids and the RBC membranes. By the strong interaction between PMB and the E. coli membrane, P-RL could attach and anchor to the E. coli; attributed to the fused RBC membrane and modified PMB, the P-RL could then efficiently neutralize both endotoxins and exotoxins from the toxin fountainhead. In vitro and in vivo results demonstrated the P-RL had a significant anchoring effect to E. coli. Moreover, compared with the existing RBC vesicles or PMB-modified liposomes, P-RL exhibited a superior therapeutic effect against RBC hemolysis, macrophage activation, and a mixed-toxin infection in mice. Potently, P-RL could inhibit E. coli O157:H7-induced skin damage, intestinal infection, and mouse death. Overall, the P-RL could potentially improve the detoxing efficiency and markedly expand the detoxification spectrum of current antivirulence systems, which provides different insights into drug-resistant E. coli treatment.
Collapse
Affiliation(s)
- Lixian Jiang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuying Zhu
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengwei Luan
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiazhen Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ge Ru
- NMPA Key Laboratory for Monitoring and Evaluation of Cosmetics, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| | - Jian-Guo Fu
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Nina Sang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Xiong
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Yuwei He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Guo-Qiang Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiange Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 311402, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
58
|
Mirzaaghasi A, Han Y, Ahn SH, Choi C, Park JH. Biodistribution and Pharmacokinectics of Liposomes and Exosomes in a Mouse Model of Sepsis. Pharmaceutics 2021; 13:427. [PMID: 33809966 PMCID: PMC8004782 DOI: 10.3390/pharmaceutics13030427] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/24/2023] Open
Abstract
Exosomes have attracted considerable attention as drug delivery vehicles because their biological properties can be utilized for selective delivery of therapeutic cargoes to disease sites. In this context, analysis of the in vivo behaviors of exosomes in a diseased state is required to maximize their therapeutic potential as drug delivery vehicles. In this study, we investigated biodistribution and pharmacokinetics of HEK293T cell-derived exosomes and PEGylated liposomes, their synthetic counterparts, into healthy and sepsis mice. We found that biodistribution and pharmacokinetics of exosomes were significantly affected by pathophysiological conditions of sepsis compared to those of liposomes. In the sepsis mice, a substantial number of exosomes were found in the lung after intravenous injection, and their prolonged blood residence was observed due to the liver dysfunction. However, liposomes did not show such sepsis-specific effects significantly. These results demonstrate that exosome-based therapeutics can be developed to manage sepsis and septic shock by virtue of their sepsis-specific in vivo behaviors.
Collapse
Affiliation(s)
- Amin Mirzaaghasi
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; (A.M.); (Y.H.)
| | - Yunho Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; (A.M.); (Y.H.)
| | - So-Hee Ahn
- Analytic Development Team, ILIAS Biologics Incorporated, Daejeon 34014, Korea;
| | - Chulhee Choi
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; (A.M.); (Y.H.)
- Analytic Development Team, ILIAS Biologics Incorporated, Daejeon 34014, Korea;
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; (A.M.); (Y.H.)
| |
Collapse
|
59
|
Leong K, Gaglani B, Khanna AK, McCurdy MT. Novel Diagnostics and Therapeutics in Sepsis. Biomedicines 2021; 9:biomedicines9030311. [PMID: 33803628 PMCID: PMC8003067 DOI: 10.3390/biomedicines9030311] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Sepsis management demands early diagnosis and timely treatment that includes source control, antimicrobial therapy, and resuscitation. Currently employed diagnostic tools are ill-equipped to rapidly diagnose sepsis and isolate the offending pathogen, which limits the ability to offer targeted and lowest-toxicity treatment. Cutting edge diagnostics and therapeutics in development may improve time to diagnosis and address two broad management principles: (1) source control by removing the molecular infectious stimulus of sepsis, and (2) attenuation of the pathological immune response allowing the body to heal. This review addresses novel diagnostics and therapeutics and their role in the management of sepsis.
Collapse
Affiliation(s)
- Kieran Leong
- Division of Pulmonary & Critical Care, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Bhavita Gaglani
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University Hospital, Winston-Salem, NC 27157, USA; (B.G.); (A.K.K.)
| | - Ashish K. Khanna
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University Hospital, Winston-Salem, NC 27157, USA; (B.G.); (A.K.K.)
- Department of Outcomes Research, Outcomes Research Consortium, Cleveland, OH 44195, USA
| | - Michael T. McCurdy
- Division of Pulmonary & Critical Care, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Correspondence:
| |
Collapse
|
60
|
Besançon H, Babiychuk V, Larpin Y, Köffel R, Schittny D, Brockhus L, Hathaway LJ, Sendi P, Draeger A, Babiychuk E. Tailored liposomal nanotraps for the treatment of Streptococcal infections. J Nanobiotechnology 2021; 19:46. [PMID: 33588835 PMCID: PMC7885208 DOI: 10.1186/s12951-021-00775-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/11/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Streptococcal infections are associated with life-threatening pneumonia and sepsis. The rise in antibiotic resistance calls for novel approaches to treat bacterial diseases. Anti-virulence strategies promote a natural way of pathogen clearance by eliminating the advantage provided to bacteria by their virulence factors. In contrast to antibiotics, anti-virulence agents are less likely to exert selective evolutionary pressure, which is a prerequisite for the development of drug resistance. As part of their virulence mechanism, many bacterial pathogens secrete cytolytic exotoxins (hemolysins) that destroy the host cell by destabilizing their plasma membrane. Liposomal nanotraps, mimicking plasmalemmal structures of host cells that are specifically targeted by bacterial toxins are being developed in order to neutralize-by competitive sequestration-numerous exotoxins. RESULTS In this study, the liposomal nanotrap technology is further developed to simultaneously neutralize the whole palette of cytolysins produced by Streptococcus pneumoniae, Streptococcus pyogenes and Streptococcus dysgalactiae subspecies equisimilis-pathogens that can cause life-threatening streptococcal toxic shock syndrome. We show that the mixture of liposomes containing high amounts of cholesterol and liposomes composed exclusively of choline-containing phospholipids is fully protective against the combined action of exotoxins secreted by these pathogens. CONCLUSIONS Unravelling the universal mechanisms that define targeting of host cells by streptococcal cytolysins paves the way for a broad-spectrum anti-toxin therapy that can be applied without a diagnostic delay for the treatment of bacterial infections including those caused by antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Hervé Besançon
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | | | - Yu Larpin
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - René Köffel
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Dominik Schittny
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Lara Brockhus
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, University of Bern, 3001, Bern, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, 3001, Bern, Switzerland
| | - Annette Draeger
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Eduard Babiychuk
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland.
| |
Collapse
|
61
|
Yang D. Application of Nanotechnology in the COVID-19 Pandemic. Int J Nanomedicine 2021; 16:623-649. [PMID: 33531805 PMCID: PMC7847377 DOI: 10.2147/ijn.s296383] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by SARS-CoV-2 infection, has been prevalent worldwide for almost a year. In early 2000, there was an outbreak of SARS-CoV, and in early 2010, a similar dissemination of infection by MERS-CoV occurred. However, no clear explanation for the spread of SARS-CoV-2 and a massive increase in the number of infections has yet been proposed. The best solution to overcome this pandemic is the development of suitable and effective vaccines and therapeutics. Fortunately, for SARS-CoV-2, the genome sequence and protein structure have been published in a short period, making research and development for prevention and treatment relatively easy. In addition, intranasal drug delivery has proven to be an effective method of administration for treating viral lung diseases. In recent years, nanotechnology-based drug delivery systems have been applied to intranasal drug delivery to overcome various limitations that occur during mucosal administration, and advances have been made to the stage where effective drug delivery is possible. This review describes the accumulated knowledge of the previous SARS-CoV and MERS-CoV infections and aims to help understand the newly emerged SARS-CoV-2 infection. Furthermore, it elucidates the achievements in developing COVID-19 vaccines and therapeutics to date through existing approaches. Finally, the applicable nanotechnology approach is described in detail, and vaccines and therapeutic drugs developed based on nanomedicine, which are currently undergoing clinical trials, have presented the potential to become innovative alternatives for overcoming COVID-19.
Collapse
Affiliation(s)
- Dongki Yang
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
62
|
Ford CA, Hurford IM, Cassat JE. Antivirulence Strategies for the Treatment of Staphylococcus aureus Infections: A Mini Review. Front Microbiol 2021; 11:632706. [PMID: 33519793 PMCID: PMC7840885 DOI: 10.3389/fmicb.2020.632706] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium capable of infecting nearly all host tissues, causing severe morbidity and mortality. Widespread antimicrobial resistance has emerged among S. aureus clinical isolates, which are now the most frequent causes of nosocomial infection among drug-resistant pathogens. S. aureus produces an array of virulence factors that enhance in vivo fitness by liberating nutrients from the host or evading host immune responses. Staphylococcal virulence factors have been identified as viable therapeutic targets for treatment, as they contribute to disease pathogenesis, tissue injury, and treatment failure. Antivirulence strategies, or treatments targeting virulence without direct toxicity to the inciting pathogen, show promise as an adjunctive therapy to traditional antimicrobials. This Mini Review examines recent research on S. aureus antivirulence strategies, with an emphasis on translational studies. While many different virulence factors have been investigated as therapeutic targets, this review focuses on strategies targeting three virulence categories: pore-forming toxins, immune evasion mechanisms, and the S. aureus quorum sensing system. These major areas of S. aureus antivirulence research demonstrate broad principles that may apply to other human pathogens. Finally, challenges of antivirulence research are outlined including the potential for resistance, the need to investigate multiple infection models, and the importance of studying antivirulence in conjunction with traditional antimicrobial treatments.
Collapse
Affiliation(s)
- Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Ian M. Hurford
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James E. Cassat
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
63
|
Aguilera-Correa JJ, Esteban J, Vallet-Regí M. Inorganic and Polymeric Nanoparticles for Human Viral and Bacterial Infections Prevention and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:E137. [PMID: 33435597 PMCID: PMC7826792 DOI: 10.3390/nano11010137] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Infectious diseases hold third place in the top 10 causes of death worldwide and were responsible for more than 6.7 million deaths in 2016. Nanomedicine is a multidisciplinary field which is based on the application of nanotechnology for medical purposes and can be defined as the use of nanomaterials for diagnosis, monitoring, control, prevention, and treatment of diseases, including infectious diseases. One of the most used nanomaterials in nanomedicine are nanoparticles, particles with a nano-scale size that show highly tunable physical and optical properties, and the capacity to a wide library of compounds. This manuscript is intended to be a comprehensive review of the available recent literature on nanoparticles used for the prevention and treatment of human infectious diseases caused by different viruses, and bacteria from a clinical point of view by basing on original articles which talk about what has been made to date and excluding commercial products, but also by highlighting what has not been still made and some clinical concepts that must be considered for futures nanoparticles-based technologies applications.
Collapse
Affiliation(s)
- John Jairo Aguilera-Correa
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Research Institute Hospital 12 de Octubre (i+12), Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, Av. Reyes Católicos 2, 28040 Madrid, Spain;
| | - María Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Research Institute Hospital 12 de Octubre (i+12), Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
64
|
Zhang S, Lu X, Wang B, Zhang G, Liu M, Geng S, Sun L, An J, Zhang Z, Zhang H. A soft anti-virulence liposome realizing the explosive release of antibiotics at an infectious site to improve antimicrobial therapy. J Mater Chem B 2021; 9:147-158. [PMID: 33226396 DOI: 10.1039/d0tb02255a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pore-forming toxins (PFTs), the most common virulence proteins, are promising therapeutic keys in bacterial infections. CAL02, consisting of sphingomyelin (Sm) and containing a maximum ratio of cholesterol (Ch), has been applied to sequester PFTs. However, Sm, a saturated phospholipid, leads to structural rigidity of the liposome, which does not benefit PFT combination. Therefore, in order to decrease the membrane rigidity and improve the fluidity of liposomes, we have introduced an unsaturated phospholipid, phosphatidylcholine (Pc), to the saturated Sm. In this report, a soft nanoliposome (called CSPL), composed of Ch, Sm and Pc, was artificially prepared. In order to further improve its antibacterial effect, vancomycin (Van) was loaded into the hydrophilic core of CSPL, where Van can be released radically at the infectious site through transmembrane pores formed by the PFTs in CSPL. This soft Van@CSPL nanoliposome with detoxification/drug release was able to inhibit the possibility of antibiotic resistance and could play a better role in treating severe invasive infections in mice.
Collapse
Affiliation(s)
- Shudong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, No. 100, Kexue Avenue, Zhengzhou 450001, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Cools F, Delputte P, Cos P. The search for novel treatment strategies for Streptococcus pneumoniae infections. FEMS Microbiol Rev 2021; 45:6064299. [PMID: 33399826 PMCID: PMC8371276 DOI: 10.1093/femsre/fuaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/01/2021] [Indexed: 12/13/2022] Open
Abstract
This review provides an overview of the most important novel treatment strategies against Streptococcus pneumoniae infections published over the past 10 years. The pneumococcus causes the majority of community-acquired bacterial pneumonia cases, and it is one of the prime pathogens in bacterial meningitis. Over the last 10 years, extensive research has been conducted to prevent severe pneumococcal infections, with a major focus on (i) boosting the host immune system and (ii) discovering novel antibacterials. Boosting the immune system can be done in two ways, either by actively modulating host immunity, mostly through administration of selective antibodies, or by interfering with pneumococcal virulence factors, thereby supporting the host immune system to effectively overcome an infection. While several of such experimental therapies are promising, few have evolved to clinical trials. The discovery of novel antibacterials is hampered by the high research and development costs versus the relatively low revenues for the pharmaceutical industry. Nevertheless, novel enzymatic assays and target-based drug design, allow the identification of targets and the development of novel molecules to effectively treat this life-threatening pathogen.
Collapse
Affiliation(s)
- F Cools
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
66
|
Rombauts A, Abelenda-Alonso G, Cuervo G, Gudiol C, Carratalà J. Role of the inflammatory response in community-acquired pneumonia: clinical implications. Expert Rev Anti Infect Ther 2021; 20:1261-1274. [PMID: 33034228 DOI: 10.1080/14787210.2021.1834848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite adequate antibiotic coverage, community-acquired pneumonia (CAP) remains a leading cause of hospitalization and mortality worldwide. It induces both a local pulmonary and a systemic inflammatory response, particularly significant in severe cases. The intensity of the dysregulated host response varies from patient to patient and has a negative impact on survival and other outcomes. AREAS COVERED This comprehensive review summarizes the pathophysiological aspects of the inflammatory response in CAP, briefly discusses the usefulness of biomarkers, and assesses the clinical evidence for modulating the inflammatory pathways. We searched PubMed for the most relevant studies, reviews, and meta-analysis until August 2020. EXPERT OPINION Notable efforts have been made to identify biomarkers that can accurately differentiate between viral and bacterial etiology, and indeed, to enhance risk stratification in CAP. However, none has proven ideal and no recommended biomarker-guided algorithms exist. Biomarker signatures from proteomic and metabolomic studies could be more useful for such assessments. To date, most studies have produced contradictory results concerning the role of immunomodulatory agents (e.g. corticosteroids, macrolides, and statins) in CAP. Adequately identifying the population who may benefit most from effective modulation of the inflammatory response remains a challenge.
Collapse
Affiliation(s)
- Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Guillermo Cuervo
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain.,Institut Català d'Oncologia (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
67
|
Papafilippou L, Claxton A, Dark P, Kostarelos K, Hadjidemetriou M. Nanotools for Sepsis Diagnosis and Treatment. Adv Healthc Mater 2021; 10:e2001378. [PMID: 33236524 PMCID: PMC11469323 DOI: 10.1002/adhm.202001378] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Sepsis is one of the leading causes of death worldwide with high mortality rates and a pathological complexity hindering early and accurate diagnosis. Today, laboratory culture tests are the epitome of pathogen recognition in sepsis. However, their consistency remains an issue of controversy with false negative results often observed. Clinically used blood markers, C reactive protein (CRP) and procalcitonin (PCT) are indicators of an acute-phase response and thus lack specificity, offering limited diagnostic efficacy. In addition to poor diagnosis, inefficient drug delivery and the increasing prevalence of antibiotic-resistant microorganisms constitute significant barriers in antibiotic stewardship and impede effective therapy. These challenges have prompted the exploration for alternative strategies that pursue accurate diagnosis and effective treatment. Nanomaterials are examined for both diagnostic and therapeutic purposes in sepsis. The nanoparticle (NP)-enabled capture of sepsis causative agents and/or sepsis biomarkers in biofluids can revolutionize sepsis diagnosis. From the therapeutic point of view, currently existing nanoscale drug delivery systems have proven to be excellent allies in targeted therapy, while many other nanotherapeutic applications are envisioned. Herein, the most relevant applications of nanomedicine for the diagnosis, prognosis, and treatment of sepsis is reviewed, providing a critical assessment of their potentiality for clinical translation.
Collapse
Affiliation(s)
- Lana Papafilippou
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
| | - Andrew Claxton
- Department of Critical CareSalford Royal Foundation TrustStott LaneSalfordM6 8HDUK
| | - Paul Dark
- Manchester NIHR Biomedical Research CentreDivision of InfectionImmunity and Respiratory MedicineUniversity of ManchesterManchesterM13 9PTUK
| | - Kostas Kostarelos
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)Campus UABBellaterraBarcelona08193Spain
| | - Marilena Hadjidemetriou
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
| |
Collapse
|
68
|
Potter M, Najer A, Klöckner A, Zhang S, Holme MN, Nele V, Che J, Massi L, Penders J, Saunders C, Doutch JJ, Edwards AM, Ces O, Stevens MM. Controlled Dendrimersome Nanoreactor System for Localized Hypochlorite-Induced Killing of Bacteria. ACS NANO 2020; 14:17333-17353. [PMID: 33290039 PMCID: PMC7760217 DOI: 10.1021/acsnano.0c07459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/23/2020] [Indexed: 05/20/2023]
Abstract
Antibiotic resistance is a serious global health problem necessitating new bactericidal approaches such as nanomedicines. Dendrimersomes (DSs) have recently become a valuable alternative nanocarrier to polymersomes and liposomes due to their molecular definition and synthetic versatility. Despite this, their biomedical application is still in its infancy. Inspired by the localized antimicrobial function of neutrophil phagosomes and the versatility of DSs, a simple three-component DS-based nanoreactor with broad-spectrum bactericidal activity is presented. This was achieved by encapsulation of glucose oxidase (GOX) and myeloperoxidase (MPO) within DSs (GOX-MPO-DSs), self-assembled from an amphiphilic Janus dendrimer, that possesses a semipermeable membrane. By external addition of glucose to GOX-MPO-DS, the production of hypochlorite (-OCl), a highly potent antimicrobial, by the enzymatic cascade was demonstrated. This cascade nanoreactor yielded a potent bactericidal effect against two important multidrug resistant pathogens, Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa), not observed for H2O2 producing nanoreactors, GOX-DS. The production of highly reactive species such as -OCl represents a harsh bactericidal approach that could also be cytotoxic to mammalian cells. This necessitates the development of strategies for activating -OCl production in a localized manner in response to a bacterial stimulus. One option of locally releasing sufficient amounts of substrate using a bacterial trigger (released toxins) was demonstrated with lipidic glucose-loaded giant unilamellar vesicles (GUVs), envisioning, e.g., implant surface modification with nanoreactors and GUVs for localized production of bactericidal agents in the presence of bacterial growth.
Collapse
Affiliation(s)
- Michael Potter
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Chemistry and Institute of Chemical Biology, Imperial College London, Molecular Sciences Research Hub, London W12 0BZ, U.K.
| | - Adrian Najer
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Anna Klöckner
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, U.K.
| | - Shaodong Zhang
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Margaret N. Holme
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Valeria Nele
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Junyi Che
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Lucia Massi
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Jelle Penders
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Catherine Saunders
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - James J. Doutch
- Rutherford
Appleton Laboratory, ISIS Neutron and Muon
Source, STFC, Didcot OX11 ODE, U.K.
| | - Andrew M. Edwards
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, U.K.
| | - Oscar Ces
- Department
of Chemistry and Institute of Chemical Biology, Imperial College London, Molecular Sciences Research Hub, London W12 0BZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
69
|
Ebbers M, Hemmer CJ, Müller-Hilke B, Reisinger EC. Immunotherapy and vaccination against infectious diseases. Wien Klin Wochenschr 2020; 133:714-720. [PMID: 33326055 PMCID: PMC7738774 DOI: 10.1007/s00508-020-01746-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022]
Abstract
Due to the overuse of antibiotics, infections, in particular those caused by multidrug-resistant bacteria, are becoming more and more frequent. Despite the worldwide introduction of antibiotic therapy, vaccines and constant improvements in hygiene, the burden of multidrug-resistant bacterial infections is increasing and is expected to rise in the future. The development of monoclonal therapeutic antibodies and specific immunomodulatory drugs represent new treatment options in the fight against infectious diseases. This article provides a brief overview of recent advances in immunomodulatory therapy and other strategies in the treatment of infectious disease.
Collapse
Affiliation(s)
- Meinolf Ebbers
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.,Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Christoph J Hemmer
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Brigitte Müller-Hilke
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Emil C Reisinger
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
70
|
Sellarès-Nadal J, Burgos J, Falcó V, Almirante B. Investigational and Experimental Drugs for Community-Acquired Pneumonia: the Current Evidence. J Exp Pharmacol 2020; 12:529-538. [PMID: 33239925 PMCID: PMC7682597 DOI: 10.2147/jep.s259286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022] Open
Abstract
Community-acquired pneumonia (CAP) is a common infection with a constantly evolving etiological spectrum. This changing etiology conditions the adequate selection of optimal therapeutic regimens, both in empirical and definitive treatments. In recent years, new antimicrobials have been approved by regulatory authorities for use in CAP, although it is necessary to continue incorporating new antimicrobial agents that improve the activity profile in relation to the appearance of bacterial resistance in certain pathogens, such as pneumococcus, Staphylococcus aureus or Pseudomonas aeruginosa. Delafloxacin, omadacycline and lefamulin are the most recently approved antibiotics for CAP. These three antibiotics have shown non-inferiority to their comparators for the treatment of CAP with an excellent safety profile. However, in the 2019 ATS/IDSA guidelines, it has been considered that more information is needed to incorporate these new drugs into community-based treatment. New antimicrobials, such as solithromycin and nemonoxacin, are currently being studied in Phase III clinical trials. Both drugs have shown non-inferiority against the comparators and an acceptable safety profile; however, they have not yet been approved by the regulatory authorities. Several drugs are being tested in Phase I and II clinical trials. These include zabofloxacin, aravofloxacin, nafithromycin, TP-271, gepotidacin, radezolid, delpazolid, and CAL02. The preliminary results of these clinical trials allow us to assure that most of these drugs may play a role in the future treatment of CAP.
Collapse
Affiliation(s)
- Juilia Sellarès-Nadal
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Joaquin Burgos
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Benito Almirante
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
71
|
Westmeier D, Siemer S, Vallet C, Steinmann J, Docter D, Buer J, Knauer SK, Stauber RH. Boosting nanotoxicity to combat multidrug-resistant bacteria in pathophysiological environments. NANOSCALE ADVANCES 2020; 2:5428-5440. [PMID: 36132026 PMCID: PMC9419095 DOI: 10.1039/d0na00644k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/21/2020] [Indexed: 06/15/2023]
Abstract
Nanomaterials are promising novel antibiotics, but often ineffective. We found that nanomaterial-bacteria complex formation occurred with various nanomaterials. The bactericidal activity of NMs strongly depends on their physical binding to (multidrug-resistant) bacteria. Nanomaterials' binding and antibiotic effect was reduced by various pathophysiological biomolecule coronas strongly inhibiting their antibiotic effects. We show from analytical to in vitro to in vivo that nanomaterial-based killing could be restored by acidic pH treatments. Here, complex formation of negatively-charged, plasma corona-covered, nanomaterials with bacteria was electrostatically enhanced by reducing bacteria's negative surface charge. Employing in vivo skin infection models, acidic pH-induced complex formation was critical to counteract Staphylococcus aureus infections by silver nanomaterials. We explain why nano-antibiotics show reduced activity and provide a clinically practical solution.
Collapse
Affiliation(s)
- Dana Westmeier
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Svenja Siemer
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Cecilia Vallet
- Department of Molecular Biology II, Center for Medical Biotechnology/Nanointegration (ZMB/CENIDE), University Duisburg-Essen, Universitätsstrasse 5 45117 Essen Germany
| | - Jörg Steinmann
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen Hufelandstrasse 55 45112 Essen Germany
| | - Dominic Docter
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen Hufelandstrasse 55 45112 Essen Germany
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology/Nanointegration (ZMB/CENIDE), University Duisburg-Essen, Universitätsstrasse 5 45117 Essen Germany
| | - Roland H Stauber
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| |
Collapse
|
72
|
Rao L, Xia S, Xu W, Tian R, Yu G, Gu C, Pan P, Meng QF, Cai X, Qu D, Lu L, Xie Y, Jiang S, Chen X. Decoy nanoparticles protect against COVID-19 by concurrently adsorbing viruses and inflammatory cytokines. Proc Natl Acad Sci U S A 2020; 117:27141-27147. [PMID: 33024017 PMCID: PMC7959535 DOI: 10.1073/pnas.2014352117] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the urgent need to rapidly develop therapeutic strategies for such emerging viruses without effective vaccines or drugs. Here, we report a decoy nanoparticle against COVID-19 through a powerful two-step neutralization approach: virus neutralization in the first step followed by cytokine neutralization in the second step. The nanodecoy, made by fusing cellular membrane nanovesicles derived from human monocytes and genetically engineered cells stably expressing angiotensin converting enzyme II (ACE2) receptors, possesses an antigenic exterior the same as source cells. By competing with host cells for virus binding, these nanodecoys effectively protect host cells from the infection of pseudoviruses and authentic SARS-CoV-2. Moreover, relying on abundant cytokine receptors on the surface, the nanodecoys efficiently bind and neutralize inflammatory cytokines including interleukin 6 (IL-6) and granulocyte-macrophage colony-stimulating factor (GM-CSF), and significantly suppress immune disorder and lung injury in an acute pneumonia mouse model. Our work presents a simple, safe, and robust antiviral nanotechnology for ongoing COVID-19 and future potential epidemics.
Collapse
Affiliation(s)
- Lang Rao
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Shuai Xia
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Wei Xu
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Chenjian Gu
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Pan Pan
- Institute of Medical Microbiology, Jinan University, 510632 Guangzhou, China
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, 430072 Wuhan, China
| | - Qian-Fang Meng
- School of Physics and Technology, Wuhan University, 430072 Wuhan, China
| | - Xia Cai
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Di Qu
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Lu Lu
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China;
| | - Youhua Xie
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China;
| | - Shibo Jiang
- Biosafety Level 3 Laboratory, Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China;
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
73
|
Gheorghita Puscaselu R, Lobiuc A, Dimian M, Covasa M. Alginate: From Food Industry to Biomedical Applications and Management of Metabolic Disorders. Polymers (Basel) 2020; 12:E2417. [PMID: 33092194 PMCID: PMC7589871 DOI: 10.3390/polym12102417] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Initially used extensively as an additive and ingredient in the food industry, alginate has become an important compound for a wide range of industries and applications, such as the medical, pharmaceutical and cosmetics sectors. In the food industry, alginate has been used to coat fruits and vegetables, as a microbial and viral protection product, and as a gelling, thickening, stabilizing or emulsifying agent. Its biocompatibility, biodegradability, nontoxicity and the possibility of it being used in quantum satis doses prompted scientists to explore new properties for alginate usage. Thus, the use of alginate has been expanded so as to be directed towards the pharmaceutical and biomedical industries, where studies have shown that it can be used successfully as biomaterial for wound, hydrogel, and aerogel dressings, among others. Furthermore, the ability to encapsulate natural substances has led to the possibility of using alginate as a drug coating and drug delivery agent, including the encapsulation of probiotics. This is important considering the fact that, until recently, encapsulation and coating agents used in the pharmaceutical industry were limited to the use of lactose, a potentially allergenic agent or gelatin. Obtained at a relatively low cost from marine brown algae, this hydrocolloid can also be used as a potential tool in the management of diabetes, not only as an insulin delivery agent but also due to its ability to improve insulin resistance, attenuate chronic inflammation and decrease oxidative stress. In addition, alginate has been recognized as a potential weight loss treatment, as alginate supplementation has been used as an adjunct treatment to energy restriction, to enhance satiety and improve weight loss in obese individuals. Thus, alginate holds the promise of an effective product used in the food industry as well as in the management of metabolic disorders such as diabetes and obesity. This review highlights recent research advances on the characteristics of alginate and brings to the forefront the beneficial aspects of using alginate, from the food industry to the biomedical field.
Collapse
Affiliation(s)
- Roxana Gheorghita Puscaselu
- Department of Health and Human Development, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.G.P.); (A.L.)
| | - Andrei Lobiuc
- Department of Health and Human Development, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.G.P.); (A.L.)
| | - Mihai Dimian
- Department of Computers, Electronics and Automation, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
- Integrated Center for Research, Development and Innovation in Advanced Materials, Nanotechnologies, and Distributed Systems for Fabrication and Control, Stefan cel Mare University of Suceava, 720229 Suceava, Romania
| | - Mihai Covasa
- Department of Health and Human Development, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.G.P.); (A.L.)
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
74
|
Subramanian K, Iovino F, Tsikourkitoudi V, Merkl P, Ahmed S, Berry SB, Aschtgen MS, Svensson M, Bergman P, Sotiriou GA, Henriques-Normark B. Mannose receptor-derived peptides neutralize pore-forming toxins and reduce inflammation and development of pneumococcal disease. EMBO Mol Med 2020; 12:e12695. [PMID: 32985105 PMCID: PMC7645366 DOI: 10.15252/emmm.202012695] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 12/30/2022] Open
Abstract
Cholesterol‐dependent cytolysins (CDCs) are essential virulence factors for many human pathogens like Streptococcus pneumoniae (pneumolysin, PLY), Streptococcus pyogenes (streptolysin O, SLO), and Listeria monocytogenes (Listeriolysin, LLO) and induce cytolysis and inflammation. Recently, we identified that pneumococcal PLY interacts with the mannose receptor (MRC‐1) on specific immune cells thereby evoking an anti‐inflammatory response at sublytic doses. Here, we identified the interaction sites between MRC‐1 and CDCs using computational docking. We designed peptides from the CTLD4 domain of MRC‐1 that binds to PLY, SLO, and LLO, respectively. In vitro, the peptides blocked CDC‐induced cytolysis and inflammatory cytokine production by human macrophages. Also, they reduced PLY‐induced damage of the epithelial barrier integrity as well as blocked bacterial invasion into the epithelium in a 3D lung tissue model. Pre‐treatment of human DCs with peptides blocked bacterial uptake via MRC‐1 and reduced intracellular bacterial survival by targeting bacteria to autophagosomes. In order to use the peptides for treatment in vivo, we developed calcium phosphate nanoparticles (CaP NPs) as peptide nanocarriers for intranasal delivery of peptides and enhanced bioactivity. Co‐administration of peptide‐loaded CaP NPs during infection improved survival and bacterial clearance in both zebrafish and mice models of pneumococcal infection. We suggest that MRC‐1 peptides can be employed as adjunctive therapeutics with antibiotics to treat bacterial infections by countering the action of CDCs.
Collapse
Affiliation(s)
- Karthik Subramanian
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Federico Iovino
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Vasiliki Tsikourkitoudi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Padryk Merkl
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sultan Ahmed
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Samuel B Berry
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | | | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.,The Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Georgios A Sotiriou
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Lee Kong Chian School of Medicine (LKC) and Singapore Centre on Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
75
|
Feldman C, Anderson R. Platelets and Their Role in the Pathogenesis of Cardiovascular Events in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:577303. [PMID: 33042161 PMCID: PMC7527494 DOI: 10.3389/fimmu.2020.577303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains an important cause of morbidity and mortality throughout the world with much recent and ongoing research focused on the occurrence of cardiovascular events (CVEs) during the infection, which are associated with adverse short-term and long-term survival. Much of the research directed at unraveling the pathogenesis of these events has been undertaken in the settings of experimental and clinical CAP caused by the dangerous, bacterial respiratory pathogen, Streptococcus pneumoniae (pneumococcus), which remains the most common bacterial cause of CAP. Studies of this type have revealed that although platelets play an important role in host defense against infection, there is also increasing recognition that hyperactivation of these cells contributes to a pro-inflammatory, prothrombotic systemic milieu that contributes to the etiology of CVEs. In the case of the pneumococcus, platelet-driven myocardial damage and dysfunction is exacerbated by the direct cardiotoxic actions of pneumolysin, a major pore-forming toxin of this pathogen, which also acts as potent activator of platelets. This review is focused on the role of platelets in host defense against infection, including pneumococcal infection in particular, and reviews the current literature describing the potential mechanisms by which platelet activation contributes to cardiovascular complications in CAP. This is preceded by an evaluation of the burden of pneumococcal infection in CAP, the clinical features and putative pathogenic mechanisms of the CVE, and concludes with an evaluation of the potential utility of the anti-platelet activity of macrolides and various adjunctive therapies.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, Institute of Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
76
|
Liao Y, Zhang Y, Blum NT, Lin J, Huang P. Biomimetic hybrid membrane-based nanoplatforms: synthesis, properties and biomedical applications. NANOSCALE HORIZONS 2020; 5:1293-1302. [PMID: 32608425 DOI: 10.1039/d0nh00267d] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The rapid clearance and capture by the immune system pose a big challenge in targeted drug delivery using nanocarriers. Cell membrane coating endows nanoplatforms with prolonged blood circulation, enhanced immune escape, and improved targeting capability. However, monotypic cell membrane fails to meet the omnifarious needs of biomedical applications. The combination of different types of cell membranes provides a promising solution to provide multifunctional biomimetic nanoplatforms. In this review, we first discuss the feasibility of constructing biomimetic hybrid membranes and summarize current methods of preparing biomimetic hybrid membrane-based nanoplatforms (BHMNs) and their biomedical applications including drug delivery, cancer detection, detoxification, and cancer vaccines. Finally, the prospects and challenges of utilizing BHMNs for personalized medicine are also discussed.
Collapse
Affiliation(s)
- Yunyan Liao
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China.
| | | | | | | | | |
Collapse
|
77
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
78
|
Cools F, Triki D, Geerts N, Delputte P, Fourches D, Cos P. In vitro and in vivo Evaluation of in silico Predicted Pneumococcal UDPG:PP Inhibitors. Front Microbiol 2020; 11:1596. [PMID: 32760374 PMCID: PMC7373766 DOI: 10.3389/fmicb.2020.01596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/18/2020] [Indexed: 11/25/2022] Open
Abstract
Pneumonia, of which Streptococcus pneumoniae is the most common causative agent, is considered one of the three top leading causes of death worldwide. As seen in other bacterial species, antimicrobial resistance is on the rise for this pathogen. Therefore, there is a pressing need for novel antimicrobial strategies to combat these infections. Recently, uridine diphosphate glucose pyrophosphorylase (UDPG:PP) has been put forward as a potential drug target worth investigating. Moreover, earlier research demonstrated that streptococci lacking a functional galU gene (encoding for UDPG:PP) were characterized by significantly reduced in vitro and in vivo virulence. Therefore, in this study we evaluated the anti-virulence activity of potential UDPG:PP inhibitors. They were selected in silico using a tailor-made streptococcal homology model, based on earlier listerial research. While the compounds didn’t affect bacterial growth, nor affected in vitro adhesion to and phagocytosis in macrophages, the amount of polysaccharide capsule was significantly reduced after co-incubation with these inhibitors. Moreover, co-incubation proved to have a positive effect on survival in an in vivo Galleria mellonella larval infection model. Therefore, rather than targeting bacterial survival directly, these compounds proved to have an effect on streptococcal virulence by lowering the amount of polysaccharide and thereby probably boosting recognition of this pathogen by the innate immune system. While the compounds need adaptation to broaden their activity to more streptococcal strains rather than being strain-specific, this study consolidates UDPG:PP as a potential novel drug target.
Collapse
Affiliation(s)
- Freya Cools
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Dhoha Triki
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Nele Geerts
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Peter Delputte
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Denis Fourches
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Paul Cos
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
79
|
Rabanel JM, Delbreil P, Banquy X, Brambilla D, Ramassamy C. Periphery-confined particulate systems for the management of neurodegenerative diseases and toxicity: Avoiding the blood-brain-barrier challenge. J Control Release 2020; 322:286-299. [PMID: 32243978 DOI: 10.1016/j.jconrel.2020.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/07/2023]
Abstract
The blood-brain barrier prevents passage of large and hydrophilic molecules, undermining efforts to deliver most active molecules, proteins and other macromolecules. To date, nanoparticle-assisted delivery has been extensively studied to overcome this challenge but with limited success. On the other hand, for certain brain therapeutic applications, periphery-confined particles could be of immediate therapeutic usefulness. The modulation of CNS dysfunctions from the peripheral compartment is a promising approach, as it does not involve invasive interventions. From recent studies, three main roles could be identified for periphery-confined particles: brain tissue detoxification via the "sink-effect"; a "circulating drug-reservoir" effect to improve drug delivery to brain tissues, and finally, brain vascular endothelium targeting to diagnose or heal vascular-related dysfunctions. These applications are much easier to implement as they do not involve complex therapeutic and targeting strategies and do not require crossing biological barriers. Micro/nano-devices required for such applications will likely be simpler to synthesize and will involve fewer complex materials. Moreover, peripheral particles are expected to be less prone to neurotoxicity and issues related to their diffusion in confined space.
Collapse
Affiliation(s)
- Jean-Michel Rabanel
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Philippe Delbreil
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Davide Brambilla
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| |
Collapse
|
80
|
Abstract
When antibiotics are administered, orally or intravenously, they pass through different organs and layers of tissue on their way to the site of infection; this can cause dilution and/or intoxication. To overcome these problems, drug delivery vehicles have been used to encapsulate and deliver antibiotics, improving their therapeutic index while minimizing their adverse effects. Liposomes are self-assembled lipid vesicles made from at least one bilayer of phospholipids with an inner aqueous compartment. Liposomes are attractive vehicles to deliver antibiotics because they can encapsulate both hydrophobic and hydrophilic antibiotics, they have low toxicity, and they can change the biodistribution of the drug. Furthermore, liposomes have been approved by regulatory agencies. However, most developmental and mechanistic research in the field has been focused on encapsulation and delivery of anticancer drugs, a class of molecules that differ significantly in chemistry from antibiotics. In this critical Review, we discuss the state of knowledge regarding the design of liposomes for encapsulation and delivery of antibiotics and offer insight into the challenges and promises of using liposomes for antibiotic delivery.
Collapse
Affiliation(s)
- Azucena Gonzalez Gomez
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
| | - Zeinab Hosseinidoust
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L98 4LS, Canada
| |
Collapse
|
81
|
Jacques LC, Panagiotou S, Baltazar M, Senghore M, Khandaker S, Xu R, Bricio-Moreno L, Yang M, Dowson CG, Everett DB, Neill DR, Kadioglu A. Increased pathogenicity of pneumococcal serotype 1 is driven by rapid autolysis and release of pneumolysin. Nat Commun 2020; 11:1892. [PMID: 32312961 PMCID: PMC7170840 DOI: 10.1038/s41467-020-15751-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/23/2020] [Indexed: 12/17/2022] Open
Abstract
Streptococcus pneumoniae serotype 1 is the predominant cause of invasive pneumococcal disease in sub-Saharan Africa, but the mechanism behind its increased invasiveness is not well understood. Here, we use mouse models of lung infection to identify virulence factors associated with severe bacteraemic pneumonia during serotype-1 (ST217) infection. We use BALB/c mice, which are highly resistant to pneumococcal pneumonia when infected with other serotypes. However, we observe 100% mortality and high levels of bacteraemia within 24 hours when BALB/c mice are intranasally infected with ST217. Serotype 1 produces large quantities of pneumolysin, which is rapidly released due to high levels of bacterial autolysis. This leads to substantial levels of cellular cytotoxicity and breakdown of tight junctions between cells, allowing a route for rapid bacterial dissemination from the respiratory tract into the blood. Thus, our results offer an explanation for the increased invasiveness of serotype 1. The mechanisms behind the high invasiveness of Streptococcus pneumoniae serotype 1 are unclear. Here, Jacques et al. show that this feature is due to overproduction and rapid release of pneumolysin, which induces cytotoxicity and breakdown of tight junctions, allowing rapid bacterial dissemination from the respiratory tract into the blood.
Collapse
Affiliation(s)
- Laura C Jacques
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Stavros Panagiotou
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Murielle Baltazar
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | | | - Shadia Khandaker
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Rong Xu
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Laura Bricio-Moreno
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Marie Yang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | | | - Dean B Everett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi, College of Medicine, Blantyre, Malawi
| | - Daniel R Neill
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| |
Collapse
|
82
|
Beckmann N, Salyer CE, Crisologo PA, Nomellini V, Caldwell CC. Staging and Personalized Intervention for Infection and Sepsis. Surg Infect (Larchmt) 2020; 21:732-744. [PMID: 32240042 DOI: 10.1089/sur.2019.363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Sepsis is defined as a dysregulated host response to infection, resulting in life-threatening organ dysfunction. It is now understood that this dysregulation not only constitutes excessive inflammation, but also sustained immune suppression. Immune-modulatory therapies thus have great potential for novel sepsis therapies. Here, we provide a review of biomarkers and functional assays designed to immunologically stage patients with sepsis as well as therapies designed to alter the innate and adaptive immune systems of patients with sepsis beneficially. Methods: A search of PubMed/MEDLINE and clinicaltrials.gov was performed between October 1, 2019 and December 22, 2019 using search terms such as "sepsis immunotherapy," "sepsis biomarkers," "sepsis clinical trials," and variations thereof. Results: Despite more than 30 years of research, there is still no Food and Drug Administration (FDA)-cleared biomarker that has proven to be effective in either identifying patients with sepsis who are at an increased risk of adverse outcomes or responsive to specific interventions. Similarly, past clinical trials investigating new treatment strategies have rarely stratified patients with sepsis. Overall, the results of these trials have been disappointing. Novel efforts to properly gauge an individual patient's immune response and choose an appropriate immunomodulatory agent based on the results are underway. Conclusion: Our evolving understanding of the different mechanisms perturbing immune homeostasis during sepsis strongly suggests that future successes will depend on finding the right therapy for the right patient and administering it at the right time. For such a personalized medicine approach, novel biomarkers and functional assays to properly stage the patient with sepsis will be crucial. The growing repertoire of immunomodulatory agents at our disposal, as well as re-appraisal of agents that have already been tested in unstratified cohorts of patients with sepsis, may finally translate into successful treatment strategies for sepsis.
Collapse
Affiliation(s)
- Nadine Beckmann
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Christen E Salyer
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Peter A Crisologo
- Division of Podiatric Medicine and Surgery, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vanessa Nomellini
- Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| | - Charles C Caldwell
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
83
|
Abstract
Infectious diseases are a leading cause of mortality worldwide, with viruses and bacteria in particular having enormous impacts on global healthcare. One major challenge in combatting such diseases is a lack of effective drugs or specific treatments. In addition, drug resistance to currently available therapeutics and adverse effects caused by long-term overuse are both serious public health issues. A promising treatment strategy is to employ cell-membrane mimics as decoys to trap and to detain the pathogens. In this Perspective, we briefly review the infection mechanisms adopted by different pathogens at the cellular membrane interface and highlight the applications of cell-membrane-mimicking nanodecoys for systemic protection against infectious diseases. We also discuss the implication of nanodecoy-pathogen complexes in the development of vaccines. We anticipate this Perspective will provide new insights on design and development of advanced materials against emerging infectious diseases.
Collapse
Affiliation(s)
- Lang Rao
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| |
Collapse
|
84
|
Lang J, Bohn P, Bhat H, Jastrow H, Walkenfort B, Cansiz F, Fink J, Bauer M, Olszewski D, Ramos-Nascimento A, Duhan V, Friedrich SK, Becker KA, Krawczyk A, Edwards MJ, Burchert A, Huber M, Friebus-Kardash J, Göthert JR, Hardt C, Probst HC, Schumacher F, Köhrer K, Kleuser B, Babiychuk EB, Sodeik B, Seibel J, Greber UF, Lang PA, Gulbins E, Lang KS. Acid ceramidase of macrophages traps herpes simplex virus in multivesicular bodies and protects from severe disease. Nat Commun 2020; 11:1338. [PMID: 32165633 PMCID: PMC7067866 DOI: 10.1038/s41467-020-15072-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/17/2020] [Indexed: 12/20/2022] Open
Abstract
Macrophages have important protective functions during infection with herpes simplex virus type 1 (HSV-1). However, molecular mechanisms that restrict viral propagation and protect from severe disease are unclear. Here we show that macrophages take up HSV-1 via endocytosis and transport the virions into multivesicular bodies (MVBs). In MVBs, acid ceramidase (aCDase) converts ceramide into sphingosine and increases the formation of sphingosine-rich intraluminal vesicles (ILVs). Once HSV-1 particles reach MVBs, sphingosine-rich ILVs bind to HSV-1 particles, which restricts fusion with the limiting endosomal membrane and prevents cellular infection. Lack of aCDase in macrophage cultures or in Asah1-/- mice results in replication of HSV-1 and Asah1-/- mice die soon after systemic or intravaginal inoculation. The treatment of macrophages with sphingosine enhancing compounds blocks HSV-1 propagation, suggesting a therapeutic potential of this pathway. In conclusion, aCDase loads ILVs with sphingosine, which prevents HSV-1 capsids from penetrating into the cytosol.
Collapse
Affiliation(s)
- Judith Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Patrick Bohn
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Hilal Bhat
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Holger Jastrow
- Institute of Anatomy, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Institut for Experimental Immunology and Imaging, Imaging Center Essen, Electron Microscopy Unit, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Bernd Walkenfort
- Institut for Experimental Immunology and Imaging, Imaging Center Essen, Electron Microscopy Unit, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Feyza Cansiz
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Julian Fink
- Institute of Organic Chemistry, Julius-Maximilians University of Würzburg, Am Hubland, Würzburg, D-97074, Germany
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Dominik Olszewski
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Ana Ramos-Nascimento
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany
| | - Vikas Duhan
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Sarah-Kim Friedrich
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Katrin Anne Becker
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Department of Infectious Diseases, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, University Hospital Giessen and Marburg, Campus Marburg, Baldingerstr., Marburg, D-35043, Germany
| | - Magdalena Huber
- Institute of Medical Microbiology and Hospital Hygiene, Philipps-University Marburg, Hans-Meerwein Str. 2, Marburg, D-35043, Germany
| | - Justa Friebus-Kardash
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Joachim R Göthert
- Department of Hematology, West German Cancer Center, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Cornelia Hardt
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Langenbeckstr. 1, Mainz, D-55131, Germany
| | - Fabian Schumacher
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, Nuthetal, D-14558, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Heinrich-Heine-University, Universitätsstr. 1, Düsseldorf, D-40225, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, Nuthetal, D-14558, Germany
| | - Eduard B Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstr. 4, CH-3012, Bern, Switzerland
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, Julius-Maximilians University of Würzburg, Am Hubland, Würzburg, D-97074, Germany
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, Düsseldorf, D-40225, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Karl S Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.
| |
Collapse
|
85
|
Keller MD, Ching KL, Liang FX, Dhabaria A, Tam K, Ueberheide BM, Unutmaz D, Torres VJ, Cadwell K. Decoy exosomes provide protection against bacterial toxins. Nature 2020; 579:260-264. [PMID: 32132711 DOI: 10.1038/s41586-020-2066-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/09/2020] [Indexed: 12/13/2022]
Abstract
The production of pore-forming toxins that disrupt the plasma membrane of host cells is a common virulence strategy for bacterial pathogens such as methicillin-resistant Staphylococcus aureus (MRSA)1-3. It is unclear, however, whether host species possess innate immune mechanisms that can neutralize pore-forming toxins during infection. We previously showed that the autophagy protein ATG16L1 is necessary for protection against MRSA strains encoding α-toxin4-a pore-forming toxin that binds the metalloprotease ADAM10 on the surface of a broad range of target cells and tissues2,5,6. Autophagy typically involves the targeting of cytosolic material to the lysosome for degradation. Here we demonstrate that ATG16L1 and other ATG proteins mediate protection against α-toxin through the release of ADAM10 on exosomes-extracellular vesicles of endosomal origin. Bacterial DNA and CpG DNA induce the secretion of ADAM10-bearing exosomes from human cells as well as in mice. Transferred exosomes protect host cells in vitro by serving as scavengers that can bind multiple toxins, and improve the survival of mice infected with MRSA in vivo. These findings indicate that ATG proteins mediate a previously unknown form of defence in response to infection, facilitating the release of exosomes that serve as decoys for bacterially produced toxins.
Collapse
Affiliation(s)
- Matthew D Keller
- Department of Microbiology, New York University School of Medicine, New York, NY, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Krystal L Ching
- Department of Microbiology, New York University School of Medicine, New York, NY, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- Division of Advanced Research Technologies, New York University Langone Health, New York, NY, USA.,The Microscopy Labratory at New York University Langone Health, New York, NY, USA
| | - Avantika Dhabaria
- Division of Advanced Research Technologies, New York University Langone Health, New York, NY, USA.,The Proteomics Labratory at New York University Langone Health, New York, NY, USA
| | - Kayan Tam
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Beatrix M Ueberheide
- Division of Advanced Research Technologies, New York University Langone Health, New York, NY, USA.,The Proteomics Labratory at New York University Langone Health, New York, NY, USA.,The Laura and Isaac Perlmutter Cancer Center, New York, NY, USA
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY, USA.
| | - Ken Cadwell
- Department of Microbiology, New York University School of Medicine, New York, NY, USA. .,Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, USA. .,Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
86
|
Jiménez-Avalos JA, Arrevillaga-Boni G, González-López L, García-Carvajal ZY, González-Avila M. Classical methods and perspectives for manipulating the human gut microbial ecosystem. Crit Rev Food Sci Nutr 2020; 61:234-258. [PMID: 32114770 DOI: 10.1080/10408398.2020.1724075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A healthy Human Gut Microbial Ecosystem (HGME) is a necessary condition for maintaining the orderly function of the whole body. Major alterations in the normal gut microbial composition, activity and functionality (dysbiosis) by an environmental or host-related disruptive event, can compromise metabolic, inflammatory, and neurological processes, causing disorders such as obesity, inflammatory bowel disease, colorectal cancer, and depressive episodes. The restore or the maintaining of the homeostatic balance of Gut Microbiota (GM) populations (eubiosis) is possible through diet, the use of probiotics, prebiotics, antibiotics, and even Fecal Microbiota Transplantation (FMT). Although these "classic methods" represent an effective and accepted way to modulate GM, the complexity of HGME requires new approaches to control it in a more appropriate way. Among the most promising emergent strategies for modulating GM are the use of engineered nanomaterials (metallic nanoparticles (NP), polymeric-NP, quantum dots, micelles, dendrimers, and liposomes); phagotherapy (i.e., phages linked with the CRISPR/Cas9 system), and the use of antimicrobial peptides, non-antibiotic drugs, vaccines, and immunoglobulins. Here we review the current state of development, implications, advantages, disadvantages, and perspectives of the different approaches for manipulating HGME.
Collapse
Affiliation(s)
- Jorge Armando Jiménez-Avalos
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Gerardo Arrevillaga-Boni
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | | | - Zaira Yunuen García-Carvajal
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Marisela González-Avila
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| |
Collapse
|
87
|
Zou S, Wang B, Wang C, Wang Q, Zhang L. Cell membrane-coated nanoparticles: research advances. Nanomedicine (Lond) 2020; 15:625-641. [PMID: 32098564 DOI: 10.2217/nnm-2019-0388] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell membranes have been continuously imitated and used for the modification of nanoparticles (NPs) to improve NP biological properties. Cell membrane-coated NPs, where core NPs are wrapped with plasma membrane vesicles, show high biocompatibility, targeting specificity and low side effects. Compared with conventional strategies, this novel approach directly leverages intact and natural functions of cell membranes, instead of replicating these features via synthetic techniques. This top-down technique bestows NPs with enhanced biointerfacing capabilities with potential in the diagnosis and treatment of cancer, infection and other diseases. Herein, we report on the advances in cell membrane-coated NPs, including the preparation process, source cell membranes for wrapping and potential applications of these cell membrane-coated NPs.
Collapse
Affiliation(s)
- Shuaijun Zou
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Beilei Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Chao Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Qianqian Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Liming Zhang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| |
Collapse
|
88
|
Critical Parameters for the Development of Novel Therapies for Severe and Resistant Infections-A Case Study on CAL02, a Non-Traditional Broad-Spectrum Anti-Virulence Drug. Antibiotics (Basel) 2020; 9:antibiotics9020094. [PMID: 32098274 PMCID: PMC7168140 DOI: 10.3390/antibiotics9020094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/16/2020] [Accepted: 02/16/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Poor outcomes in severe and resistant infections, together with the economic struggles of companies active in the field of anti-infective development, call for new solutions and front runners with novel approaches. Among “non-traditional” approaches, blocking virulence could be a game changer. Objectives: This review offers a perspective on parameters that have determined the development path of CAL02, a novel anti-virulence agent, with a view to steering clear of the obstacles and limitations that impede market sustainability for new anti-infective drugs. Conclusions and implications of key findings: This case study highlights four pillars that may support the development of other non-traditional drugs and, concurrently, provide a new model that could reshape the field. Therapeutic triggers, study designs, and economic parameters are discussed.
Collapse
|
89
|
Koulenti D, Xu E, Song A, Sum Mok IY, Karageorgopoulos DE, Armaganidis A, Tsiodras S, Lipman J. Emerging Treatment Options for Infections by Multidrug-Resistant Gram-Positive Microorganisms. Microorganisms 2020; 8:E191. [PMID: 32019171 PMCID: PMC7074912 DOI: 10.3390/microorganisms8020191] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Antimicrobial agents are currently the mainstay of treatment for bacterial infections worldwide. However, due to the increased use of antimicrobials in both human and animal medicine, pathogens have now evolved to possess high levels of multi-drug resistance, leading to the persistence and spread of difficult-to-treat infections. Several current antibacterial agents active against Gram-positive bacteria will be rendered useless in the face of increasing resistance rates. There are several emerging antibiotics under development, some of which have been shown to be more effective with an improved safety profile than current treatment regimens against Gram-positive bacteria. We will extensively discuss these antibiotics under clinical development (phase I-III clinical trials) to combat Gram-positive bacteria, such as Staphylococcus aureus, Enterococcus faecium and Streptococcus pneumoniae. We will delve into the mechanism of actions, microbiological spectrum, and, where available, the pharmacokinetics, safety profile, and efficacy of these drugs, aiming to provide a comprehensive review to the involved stakeholders.
Collapse
Affiliation(s)
- Despoina Koulenti
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
- 2nd Critical Care Department, Attikon University Hospital, 12462 Athens, Greece;
| | - Elena Xu
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Andrew Song
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Isaac Yin Sum Mok
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Drosos E. Karageorgopoulos
- 4th Department of Internal Medicine, Attikon University Hospital, 12462 Athens, Greece; (D.E.K.); (S.T.)
| | | | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Attikon University Hospital, 12462 Athens, Greece; (D.E.K.); (S.T.)
| | - Jeffrey Lipman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
- Department of Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
- Anesthesiology and Critical Care, Centre Hospitalier Universitaire De Nîmes (CHU), University of Montpellier, 30029 Nîmes, France
| |
Collapse
|
90
|
Desikan R, Maiti PK, Ayappa KG. Predicting interfacial hot-spot residues that stabilize protein-protein interfaces in oligomeric membrane-toxin pores through hydrogen bonds and salt bridges. J Biomol Struct Dyn 2020; 39:20-34. [DOI: 10.1080/07391102.2020.1711806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Rajat Desikan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Prabal K. Maiti
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India
| | - K. Ganapathy Ayappa
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
91
|
Larpin Y, Besançon H, Iacovache MI, Babiychuk VS, Babiychuk EB, Zuber B, Draeger A, Köffel R. Bacterial pore-forming toxin pneumolysin: Cell membrane structure and microvesicle shedding capacity determines differential survival of cell types. FASEB J 2019; 34:1665-1678. [PMID: 31914676 DOI: 10.1096/fj.201901737rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/14/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022]
Abstract
Bacterial infectious diseases can lead to death or to serious illnesses. These outcomes are partly the consequence of pore-forming toxins, which are secreted by the pathogenic bacteria (eg, pneumolysin of Streptococcus pneumoniae). Pneumolysin binds to cholesterol within the plasma membrane of host cells and assembles to form trans-membrane pores, which can lead to Ca2+ influx and cell death. Membrane repair mechanisms exist that limit the extent of damage. Immune cells which are essential to fight bacterial infections critically rely on survival mechanisms after detrimental pneumolysin attacks. This study investigated the susceptibility of different immune cell types to pneumolysin. As a model system, we used the lymphoid T-cell line Jurkat, and myeloid cell lines U937 and THP-1. We show that Jurkat T cells are highly susceptible to pneumolysin attack. In contrast, myeloid THP-1 and U937 cells are less susceptible to pneumolysin. In line with these findings, human primary T cells are shown to be more susceptible to pneumolysin attack than monocytes. Differences in susceptibility to pneumolysin are due to (I) preferential binding of pneumolysin to Jurkat T cells and (II) cell type specific plasma membrane repair capacity. Myeloid cell survival is mostly dependent on Ca2+ induced expelling of damaged plasma membrane areas as microvesicles. Thus, in myeloid cells, first-line defense cells in bacterial infections, a potent cellular repair machinery ensures cell survival after pneumolysin attack. In lymphoid cells, which are important at later stages of infections, less efficient repair mechanisms and enhanced toxin binding renders the cells more sensitive to pneumolysin.
Collapse
Affiliation(s)
- Yu Larpin
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Hervé Besançon
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Mircea-Ioan Iacovache
- Laboratory of Experimental Morphology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Victoriia S Babiychuk
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Eduard B Babiychuk
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Benoît Zuber
- Laboratory of Experimental Morphology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Annette Draeger
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - René Köffel
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| |
Collapse
|
92
|
Zhang Q, Wu W, Zhang J, Xia X. Antimicrobial lipids in nano-carriers for antibacterial delivery. J Drug Target 2019; 28:271-281. [PMID: 31613147 DOI: 10.1080/1061186x.2019.1681434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Antimicrobial lipids have been recognised as broad-spectrum antibacterial agents. They can directly act on and lyse bacterial cell membrane, and inhibit bacterial growth through a range of mechanisms. Antimicrobial lipids include free fatty acids, monoglycerides, cholesteryl ester, sphingolipids and etc., with the first two being the most extensively studied. Their application is usually hindered by the low solubility of the compounds themselves, and nano-sized lipid-based carriers can endow druggability to these antimicrobial agents for they improve lipid solubility and dispersion in aqueous formulations. Nano-carriers also possess advantages in overcoming drug resistance. In this review we will discuss different kinds of antimicrobial lipids in nano-sized carriers for antibacterial delivery. CAL02 as a promising infection-controlling liposome consisted of cholesterol and sphingomyelin will also be included for it's a unique anti-infection approach, which signifies that the underlying antibacterial roles antimicrobial lipids needs to be further addressed. With the global emergence of antibiotic resistance, antimicrobial lipids formulated in nano-carriers might provide a novel alternative in combatting infectious diseases.
Collapse
Affiliation(s)
- Qianyu Zhang
- Innovative Drug Research Centre (IDRC), School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Wen Wu
- Innovative Drug Research Centre (IDRC), School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Jinqiang Zhang
- Innovative Drug Research Centre (IDRC), School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xuefeng Xia
- Innovative Drug Research Centre (IDRC), School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
93
|
The race between drug introduction and appearance of microbial resistance. Current balance and alternative approaches. Curr Opin Pharmacol 2019; 48:48-56. [DOI: 10.1016/j.coph.2019.04.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 11/13/2022]
|
94
|
Vila J, Moreno-Morales J, Ballesté-Delpierre C. Current landscape in the discovery of novel antibacterial agents. Clin Microbiol Infect 2019; 26:596-603. [PMID: 31574341 DOI: 10.1016/j.cmi.2019.09.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Standard treatments against bacterial infections are becoming ineffective due to the rise of antibacterial resistance worldwide. Classical approaches to develop new antibacterial agents are not sufficient to fulfil the current pipeline, therefore new strategies are currently being devised in the field of antibacterial discovery. OBJECTIVES The objective of this narrative review is to compile the most successful strategies for drug discovery within the antibacterial context that are currently being pursued. SOURCES Peer-reviewed publications from the MEDLINE database with robust data addressing the discovery of new antibacterial agents in the current pipeline have been selected. CONTENT Several strategies to discover new antibacterials are described in this review: (i) derivatives of known antibacterial agents; the activity of a known antimicrobial agent can be improved through two strategies: (a) the modification of the original chemical structure of an antimicrobial agent to circumvent antibacterial resistance mechanisms and (b) the development of a compound that inhibits the mechanisms of resistance to an antibacterial agent; (ii) new antibacterial agents targeting new proteins; (iii) inhibitors of virulence factors; (iv) nanoparticles; (v) antimicrobial peptides and peptidomimetics; (vi) phage therapy and enzybiotics; and (vii) antisense oligonucleotides. IMPLICATIONS This review intends to provide a positive message affirming that several different strategies to design new antibacterial agents are currently being developed, and we are therefore confident that in the near future some of the most promising approaches will come to fruition.
Collapse
Affiliation(s)
- J Vila
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic, Barcelona, Spain.
| | - J Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | | |
Collapse
|
95
|
Omersa N, Podobnik M, Anderluh G. Inhibition of Pore-Forming Proteins. Toxins (Basel) 2019; 11:E545. [PMID: 31546810 PMCID: PMC6784129 DOI: 10.3390/toxins11090545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Perforation of cellular membranes by pore-forming proteins can affect cell physiology, tissue integrity, or immune response. Since many pore-forming proteins are toxins or highly potent virulence factors, they represent an attractive target for the development of molecules that neutralize their actions with high efficacy. There has been an assortment of inhibitors developed to specifically obstruct the activity of pore-forming proteins, in addition to vaccination and antibiotics that serve as a plausible treatment for the majority of diseases caused by bacterial infections. Here we review a wide range of potential inhibitors that can specifically and effectively block the activity of pore-forming proteins, from small molecules to more specific macromolecular systems, such as synthetic nanoparticles, antibodies, antibody mimetics, polyvalent inhibitors, and dominant negative mutants. We discuss their mechanism of inhibition, as well as advantages and disadvantages.
Collapse
Affiliation(s)
- Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
96
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
97
|
Yamini G, Nestorovich EM. Multivalent Inhibitors of Channel-Forming Bacterial Toxins. Curr Top Microbiol Immunol 2019; 406:199-227. [PMID: 27469304 PMCID: PMC6814628 DOI: 10.1007/82_2016_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Rational design of multivalent molecules represents a remarkable modern tool to transform weak non-covalent interactions into strong binding by creating multiple finely-tuned points of contact between multivalent ligands and their supposed multivalent targets. Here, we describe several prominent examples where the multivalent blockers were investigated for their ability to directly obstruct oligomeric channel-forming bacterial exotoxins, such as the pore-forming bacterial toxins and B component of the binary bacterial toxins. We address problems related to the blocker/target symmetry match and nature of the functional groups, as well as chemistry and length of the linkers connecting the functional groups to their multivalent scaffolds. Using the anthrax toxin and AB5 toxin case studies, we briefly review how the oligomeric toxin components can be successfully disabled by the multivalent non-channel-blocking inhibitors, which are based on a variety of multivalent scaffolds.
Collapse
Affiliation(s)
- Goli Yamini
- Department of Biology, The Catholic University of America, Washington, D.C., 20064, USA
| | | |
Collapse
|
98
|
Seitz AP, Schumacher F, Baker J, Soddemann M, Wilker B, Caldwell CC, Gobble RM, Kamler M, Becker KA, Beck S, Kleuser B, Edwards MJ, Gulbins E. Sphingosine-coating of plastic surfaces prevents ventilator-associated pneumonia. J Mol Med (Berl) 2019; 97:1195-1211. [PMID: 31222488 PMCID: PMC6647234 DOI: 10.1007/s00109-019-01800-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/01/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Ventilator-associated pneumonia (VAP) is a major cause of morbidity and mortality in critically ill patients. Here, we employed the broad antibacterial effects of sphingosine to prevent VAP by developing a novel method of coating surfaces of endotracheal tubes with sphingosine and sphingosine analogs. Sphingosine and phytosphingosine coatings of endotracheal tubes prevent adherence and mediate killing of Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus, even in biofilms. Most importantly, sphingosine-coating of endotracheal tubes also prevented P. aeruginosa and S. aureus pneumonia in vivo. Coating of the tubes with sphingosine was stable, without obvious side effects on tracheal epithelial cells and did not induce inflammation. In summary, we describe a novel method to coat plastic surfaces and provide evidence for the application of sphingosine and phytosphingosine as novel antimicrobial coatings to prevent bacterial adherence and induce killing of pathogens on the surface of endotracheal tubes with potential to prevent biofilm formation and VAP. KEY MESSAGES: Novel dip-coating method to coat plastic surfaces with lipids. Sphingosine and phytosphingosine as novel antimicrobial coatings on plastic surface. Sphingosine coatings of endotracheal tubes prevent bacterial adherence and biofilms. Sphingosine coatings of endotracheal tubes induce killing of pathogens. Sphingosine coatings of endotracheal tubes ventilator-associated pneumonia.
Collapse
Affiliation(s)
- Aaron P Seitz
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA.
| | - Fabian Schumacher
- Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.,Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Jennifer Baker
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Matthias Soddemann
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Barbara Wilker
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Charles C Caldwell
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA.,Division of Research, Shriners Hospital for Children, Cincinnati, OH, 45229, USA
| | - Ryan M Gobble
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Markus Kamler
- Thoracic Transplantation, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Sascha Beck
- Orthopedic Surgery, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Michael J Edwards
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Erich Gulbins
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA. .,Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
99
|
Laterre PF, Colin G, Dequin PF, Dugernier T, Boulain T, Azeredo da Silveira S, Lajaunias F, Perez A, François B. CAL02, a novel antitoxin liposomal agent, in severe pneumococcal pneumonia: a first-in-human, double-blind, placebo-controlled, randomised trial. THE LANCET. INFECTIOUS DISEASES 2019; 19:620-630. [PMID: 31056427 DOI: 10.1016/s1473-3099(18)30805-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Severe community-acquired pneumonia caused by Streptococcus pneumoniae is associated with high morbidity and mortality rates. CAL02, a novel antitoxin agent with an unprecedented mode of action, consists of liposomes that capture bacterial toxins known to dysregulate inflammation, cause organ damage, and impede immune defence. We aimed to assess the safety of CAL02 as an add-on therapy to antibiotics. METHODS This randomised, double-blind, multicentre, placebo-controlled trial was done in ten intensive care units (ICUs) in France and Belgium (but only six units enrolled patients), in patients with severe community-acquired pneumococcal pneumonia who required ICU admission and had been identified as being infected with S pneumoniae. We randomly assigned participants in two stages-the first stage randomly assigned six patients (1:1) to either low-dose CAL02 or placebo, and the second stage randomly assigned 18 patients (14:4) to either high-dose CAL02 or placebo, and stratified in four blocks (4:1, 4:1, 3:1, and 3:1), in addition to standard of care. Block randomisation was done with a computer-generated random number list. Participants, investigators, other site study personnel, the sponsor, and the sponsor's designees involved in study management and monitoring were masked to the randomisation list and treatment assignment. Patients were treated with low-dose (4 mg/kg) or high-dose (16 mg/kg) CAL02 or placebo (saline), in addition to standard antibiotic therapy. Two intravenous doses of study treatment were infused, with a 24 h interval, at a concentration of 10 mg/mL, stepwise, over a maximum of 2 h on days 1 and 2. The primary objective of the study was to assess the safety and tolerability of low-dose and high-dose CAL02 in patients with severe community-acquired pneumonia treated with standard antibiotic therapy, and the primary analysis was done on the safety population (all patients who received at least one dose of the study treatment). Efficacy was a secondary outcome. This trial is registered with ClinicalTrials.gov, number NCT02583373. FINDINGS Between March 21, 2016, and Jan 13, 2018, we screened 280 patients with community-acquired pneumonia. 19 patients were enrolled and randomly assigned, resulting in 13 patients in the CAL02 groups (three assigned to low-dose CAL02 and ten assigned to high-dose CAL02) and six in the placebo group. One patient randomly assigned to placebo was allocated to the wrong treatment group and received high-dose CAL02 instead of placebo. Thus, 14 patients received CAL02 (three received low-dose CAL02 and 11 received high-dose CAL02) and five patients received placebo, constituting the safety population. At baseline, the mean APACHE II score for the total study population was 21·5 (SD 4·9; 95% CI 19·3-23·7) and 11 (58%) of 19 patients had septic shock. Adverse events occurred in 12 (86%) of 14 patients in the CAL02 treatment groups combined and all five (100%) patients in the placebo group. Serious adverse events occurred in four (29%) of 14 patients in the CAL02 treatment groups combined and two (40%) of five patients in the placebo group. One non-serious adverse event (mild increase in triglycerides) in a patient in the high-dose CAL02 group was reported as related to study drug. However, analysis of the changes in triglyceride levels in the CAL02 groups compared with the placebo group revealed no correlation with administration of CAL02. No adverse events were linked to local tolerability events. All patients, apart from one who died in the low CAL02 group (death not related to the study drug) achieved clinical cure at the test of cure visit between days 15 and 22. The sequential organ failure assessment score decreased by mean 65·0% (95% CI 50·7-79·4) in the combined CAL02 groups compared with 29·2% (12·8-45·5) in the placebo group between baseline and day 8. INTERPRETATION The nature of adverse events was consistent with the profile of the study population and CAL02 showed a promising safety profile and tolerability. However, the difference between high-dose and low-dose CAL02 could not be assessed in this study. Efficacy was in line with the expected benefits of neutralising toxins. The results of this study support further clinical development of CAL02 and provide a solid basis for a larger clinical study. FUNDING Combioxin.
Collapse
Affiliation(s)
- Pierre-François Laterre
- Intensive Care Unit, Cliniques Universitaires Saint-Luc, Université catholique de Louvain (UCL), Brussels, Belgium.
| | - Gwenhael Colin
- Medical-Surgical Intensive Care Unit, District Hospital Center, La Roche-sur-Yon, France
| | | | | | - Thierry Boulain
- Service de Médecine Intensive Réanimation, Hôpital La Source, Centre Hospitalier Régional d'Orléans, Orléans, France
| | | | | | | | - Bruno François
- Medical-Surgical Intensive Care Unit, University Hospital Limoges Dupuytren Hospital, Limoges, France; Inserm CIC-1435, University Hospital Limoges Dupuytren Hospital, Limoges, France; Inserm UMR 1092, Faculté de Médecine, Université de Limoges, Limoges, France
| |
Collapse
|
100
|
He Y, Li R, Li H, Zhang S, Dai W, Wu Q, Jiang L, Zheng Z, Shen S, Chen X, Zhu Y, Wang J, Pang Z. Erythroliposomes: Integrated Hybrid Nanovesicles Composed of Erythrocyte Membranes and Artificial Lipid Membranes for Pore-Forming Toxin Clearance. ACS NANO 2019; 13:4148-4159. [PMID: 30855941 DOI: 10.1021/acsnano.8b08964] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Pore-forming toxins (PFTs) are the most common bacterial virulence proteins and play a significant role in the pathogenesis of bacterial infections; thus, PFTs are an attractive therapeutic target in bacterial infections. Inspired by the pore-forming process and mechanism of PFTs, we designed an integrated hybrid nanovesicle-the erythroliposome (called the RM-PL)-for PFT detoxification by fusing natural red blood cell (RBC) membranes with artificial lipid membranes. The lipid and RBC membranes were mutually beneficial when integrated into a hybrid nanovesicle structure. The RBC membrane endowed RM-PLs with the capacity for detoxification, while the PEGylated lipid membrane stabilized the RM-PLs and greatly improved the detoxification capacity of the RBC membrane. With α-hemolysin (Hlα) as a model PFT, we demonstrated that RM-PLs could not only significantly reduce the toxicity of Hlα to erythrocytes in vitro but also effectively sponge Hlα in vivo and rescue mice from Hlα-induced damage. Moreover, the high detoxification capacity of RM-PLs was shown to be partly related to the expression of the Hlα receptor protein, a disintegrin and metalloproteinase domain-containing protein 10 on the RBC membrane. Consequently, as a component integrating natural and artificial materials, the erythroliposome nanoplatform inspires potential strategies for antivirulence therapy.
Collapse
Affiliation(s)
- Yuwei He
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Haichun Li
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Shuya Zhang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Wentao Dai
- Shanghai Center for Bioinformation Technology , Shanghai Industrial Technology Institute , Shanghai 201203 , China
| | - Qian Wu
- Shanghai Center for Bioinformation Technology , Shanghai Industrial Technology Institute , Shanghai 201203 , China
| | - Lixian Jiang
- Innovation Research Institute of Traditional Chinese Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Zicong Zheng
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Shun Shen
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
- Institute of Integrated Chinese and Western Medicine , Fudan University , Shanghai 200040 , China
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| |
Collapse
|