1
|
Zhu J, Wen T, Ma Y, Zeng Q, Li P, Zhou W. Biomimetic hyaluronic acid-stabilized zinc oxide nanoparticles in acne treatment: A preclinical and clinical approach. J Control Release 2025; 382:113754. [PMID: 40254135 DOI: 10.1016/j.jconrel.2025.113754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/22/2025]
Abstract
Acne vulgaris is a common chronic inflammatory skin condition, often caused by C. acnes infection. While ZnO has shown promise as an antibacterial agent in acne treatment, concerns over toxicity and non-selective bacterial killing remain. In this study we developed a hyaluronic acid-stabilized nano‑zinc oxide (HA-ZnO) formulation aimed at enhancing the therapeutic efficacy and safety of ZnO for acne treatment. HA-ZnO was synthesized through biomimetic mineralization. HA-ZnO targeted acne-prone areas, especially sebaceous glands, without skin penetration. HA-ZnO demonstrated selective antibacterial activity against C. acnes, exhibiting a killing efficacy more than 16 times greater than that against S. epidermidis. The HA coating also improved ZnO's stability in acidic conditions, mitigating potential toxicity and side effects. Additionally, the sustained release of Zn2+ promoted cell proliferation and migration, reducing sebum secretion, and exerting anti-inflammatory effects, supporting scar-free acne repair and preventing recurrence. In preclinical models, HA-ZnO outperformed erythromycin ointment in treating acne, with no toxicity observed in zebrafish and HET-CAM. A clinical trial further confirmed its efficacy in reducing acne lesions and redness, with high safety. These results highlight HA-ZnO as a promising therapeutic strategy for acne, combining potent antibacterial and skin-repairing effects with enhanced safety.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Tiao Wen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yunxiao Ma
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China; Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Qingya Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Pei Li
- Hunan BeautySci Biotech Co., Ltd, Hunan 410122, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China; Hunan BeautySci Biotech Co., Ltd, Hunan 410122, China.
| |
Collapse
|
2
|
Jiang JY, Xue D, Gong JS, Zheng QX, Zhang YS, Su C, Xu ZH, Shi JS. A comprehensive review on microbial hyaluronan-degrading enzymes: from virulence factors to biotechnological tools. BIORESOUR BIOPROCESS 2024; 11:114. [PMID: 39722064 DOI: 10.1186/s40643-024-00832-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Hyaluronan (HA), a natural high molecular weight polysaccharide, has extensive applications in cosmetology and medical treatment. Hyaluronan-degrading enzymes (Hyals) act as molecular scissors that cleave HA by breaking the glucosidic linkage. Hyals are present in diverse organisms, including vertebrates, invertebrates and microorganisms, and play momentous roles in biological processes. In recent years, microbial Hyals (mHyals) have gained considerable attention for their exceptional performance in the production and processing of HA. Moreover, the applications of mHyals have been greatly extended to various biomedical fields. To explore the potential applications of mHyals, a thorough comprehension is imperative. In this context, this review systematically summarizes the sources, structures, mechanisms and enzymatic properties of mHyals and discusses their biological functions in host invasion, disease development, and regulation of intestinal flora. Furthermore, versatile applications inspired by their biological functions in medicine development, molecular biology, and industrial biotechnology are comprehensively reviewed. Finally, prospects are presented to emphasize the importance of exploration, expression and characterization of mHyals and the necessity of excavating their potential in biotechnological fields.
Collapse
Affiliation(s)
- Jia-Yu Jiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Dai Xue
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
- Affiliated Children's Hospital of Jiangnan University, Wuxi, 214023, People's Republic of China
| | - Jin-Song Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China.
| | - Qin-Xin Zheng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Yue-Sheng Zhang
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, 2800, Kgs, Lyngby, Denmark
| | - Chang Su
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Zheng-Hong Xu
- School of Biotechnology, Jiangnan University, Wuxi, 214122, People's Republic of China
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, People's Republic of China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China.
| |
Collapse
|
3
|
Kumar M, Virmani T, Kumar G, Deshmukh R, Sharma A, Duarte S, Brandão P, Fonte P. Nanocarriers in Tuberculosis Treatment: Challenges and Delivery Strategies. Pharmaceuticals (Basel) 2023; 16:1360. [PMID: 37895831 PMCID: PMC10609727 DOI: 10.3390/ph16101360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The World Health Organization identifies tuberculosis (TB), caused by Mycobacterium tuberculosis, as a leading infectious killer. Although conventional treatments for TB exist, they come with challenges such as a heavy pill regimen, prolonged treatment duration, and a strict schedule, leading to multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains. The rise of MDR strains endangers future TB control. Despite these concerns, the hunt for an efficient treatment continues. One breakthrough has been the use of nanotechnology in medicines, presenting a novel approach for TB treatment. Nanocarriers, such as lipid nanoparticles, nanosuspensions, liposomes, and polymeric micelles, facilitate targeted delivery of anti-TB drugs. The benefits of nanocarriers include reduced drug doses, fewer side effects, improved drug solubility, better bioavailability, and improved patient compliance, speeding up recovery. Additionally, nanocarriers can be made even more targeted by linking them with ligands such as mannose or hyaluronic acid. This review explores these innovative TB treatments, including studies on nanocarriers containing anti-TB drugs and related patents.
Collapse
Affiliation(s)
- Mahesh Kumar
- School of Pharmaceutical Sciences, Modern Vidya Niketan University, Palwal 121105, India; (M.K.); (G.K.); (A.S.)
| | - Tarun Virmani
- School of Pharmaceutical Sciences, Modern Vidya Niketan University, Palwal 121105, India; (M.K.); (G.K.); (A.S.)
| | - Girish Kumar
- School of Pharmaceutical Sciences, Modern Vidya Niketan University, Palwal 121105, India; (M.K.); (G.K.); (A.S.)
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India;
| | - Ashwani Sharma
- School of Pharmaceutical Sciences, Modern Vidya Niketan University, Palwal 121105, India; (M.K.); (G.K.); (A.S.)
| | - Sofia Duarte
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisboa, 1049-001 Lisbon, Portugal; (S.D.); (P.B.)
- Associate Laboratory i4HB—Institute for Health and Bio-Economy, Instituto Superior Técnico, University of Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Pedro Brandão
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisboa, 1049-001 Lisbon, Portugal; (S.D.); (P.B.)
- Associate Laboratory i4HB—Institute for Health and Bio-Economy, Instituto Superior Técnico, University of Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, 2829-511 Almada, Portugal
- CQC-IMS, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
| | - Pedro Fonte
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisboa, 1049-001 Lisbon, Portugal; (S.D.); (P.B.)
- Associate Laboratory i4HB—Institute for Health and Bio-Economy, Instituto Superior Técnico, University of Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Center for Marine Sciences (CCMar), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
| |
Collapse
|
4
|
Zamboni F, Wong CK, Collins MN. Hyaluronic acid association with bacterial, fungal and viral infections: Can hyaluronic acid be used as an antimicrobial polymer for biomedical and pharmaceutical applications? Bioact Mater 2023; 19:458-473. [PMID: 35574061 PMCID: PMC9079116 DOI: 10.1016/j.bioactmat.2022.04.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/23/2022] [Indexed: 12/21/2022] Open
Abstract
The relationships between hyaluronic acid (HA) and pathological microorganisms incite new understandings on microbial infection, tissue penetration, disease progression and lastly, potential treatments. These understandings are important for the advancement of next generation antimicrobial therapeutical strategies for the control of healthcare-associated infections. Herein, this review will focus on the interplay between HA, bacteria, fungi, and viruses. This review will also comprehensively detail and discuss the antimicrobial activity displayed by various HA molecular weights for a variety of biomedical and pharmaceutical applications, including microbiology, pharmaceutics, and tissue engineering.
Collapse
Affiliation(s)
- Fernanda Zamboni
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| |
Collapse
|
5
|
Zheng X, Wang B, Tang X, Mao B, Zhang Q, Zhang T, Zhao J, Cui S, Chen W. Absorption, metabolism, and functions of hyaluronic acid and its therapeutic prospects in combination with microorganisms: A review. Carbohydr Polym 2023; 299:120153. [PMID: 36876779 DOI: 10.1016/j.carbpol.2022.120153] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022]
Abstract
Hyaluronic acid (HA) is key to the stability of the internal environment of tissues. HA content in tissues gradually decreases with age, causing age-related health problems. Exogenous HA supplements are used to prevent or treat these problems including skin dryness and wrinkles, intestinal imbalance, xerophthalmia, and arthritis after absorption. Moreover, some probiotics are able to promote endogenous HA synthesis and alleviate symptoms caused by HA loss, thus introducing potential preventative or therapeutic applications of HA and probiotics. Here, we review the oral absorption, metabolism, and biological function of HA as well as the potential role of probiotics and HA in increasing the efficacy of HA supplements.
Collapse
Affiliation(s)
- Xueli Zheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Botao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Bloomage Biotechnology Co., Ltd, Jinan 250000, China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Tianmeng Zhang
- Bloomage Biotechnology Co., Ltd, Jinan 250000, China; School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
6
|
Sindelar M, Jilkova J, Kubala L, Velebny V, Turkova K. Hyaluronidases and hyaluronate lyases: From humans to bacteriophages. Colloids Surf B Biointerfaces 2021; 208:112095. [PMID: 34507069 DOI: 10.1016/j.colsurfb.2021.112095] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/05/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022]
Abstract
Hyaluronan is a non-sulfated negatively-charged linear polymer distributed in most parts of the human body, where it is located around cells in the extracellular matrix of connective tissues and plays an essential role in the organization of tissue architecture. Moreover, hyaluronan is involved in many biological processes and used in many clinical, cosmetic, pharmaceutic, and biotechnological applications worldwide. As interest in hyaluronan applications increases, so does interest in hyaluronidases and hyaluronate lyases, as these enzymes play a major part in hyaluronan degradation. Many hyaluronidases and hyaluronate lyases produced by eukaryotic cells, bacteria, and bacteriophages have so far been described and annotated, and their ability to cleave hyaluronan has been experimentally proven. These enzymes belong to several carbohydrate-active enzyme families, share very low sequence identity, and differ in their cleaving mechanisms and in their structural and functional properties. This review presents a summary of annotated and characterized hyaluronidases and hyaluronate lyases isolated from different sources belonging to distinct protein families, with a main focus on the binding and catalytic residues of the discussed enzymes in the context of their biochemical properties. In addition, the application potential of individual groups of hyaluronidases and hyaluronate lyases is evaluated.
Collapse
Affiliation(s)
- Martin Sindelar
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, 61265, Brno, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Jana Jilkova
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic; Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Lukas Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, 61265, Brno, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 65691, Brno, Czech Republic
| | - Vladimir Velebny
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| | - Kristyna Turkova
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, 61265, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 65691, Brno, Czech Republic.
| |
Collapse
|
7
|
Simonson AW, Umstead TM, Lawanprasert A, Klein B, Almarzooqi S, Halstead ES, Medina SH. Extracellular matrix-inspired inhalable aerogels for rapid clearance of pulmonary tuberculosis. Biomaterials 2021; 273:120848. [PMID: 33915409 DOI: 10.1016/j.biomaterials.2021.120848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/31/2022]
Abstract
Tuberculosis (TB) remains a leading cause of death from a single infectious agent, and limiting the spread of multidrug-resistant TB (MDR-TB) is now an urgent global health priority. Essential to the persistence of this disease is the ability of Mycobacterium tuberculosis (Mtb) to circumvent host defenses by infecting lung macrophages to create a cellular niche for its survival and proliferation. This has urged the development of new therapeutic strategies that act through mechanisms distinct from conventional antibiotics, and thus are effective against MDR bacteria, while being able to efficiently kill persister Mtb cells in infected host macrophages. Here, we report a new class of gel-like microparticle aerosols, or 'aerogels', designed to exploit metabolic vulnerabilities of Mtb pathogens and TB-infected macrophages to enable preferential delivery of synergistic peptide-antibiotic combinations for potent and rapid antitubercular therapy. This is achieved by formulating aerogels through the supramolecular assembly of a de novo designed anti-TB peptide and the extracellular matrix (ECM)-derived polysaccharide, hyaluronic acid (HA). Importantly, HA serves as a nutrient source for Mtb cells during tissue invasion and proliferation, and is recognized by CD44 receptors highly expressed on lung macrophages during TB infection. By exploiting this metabolic substrate for pathogen targeting, HA aerogels are shown to avidly bind and kill both drug-sensitive and drug-resistant mycobacteria, while being efficiently internalized into macrophage host cells in vitro and in vivo to clear Mtb persisters. This multifaceted bioactivity suggests aerogels may serve as a versatile inhalable platform upon which novel biomaterials-enabled therapeutics can be developed to rapidly clear pulmonary MDR-TB.
Collapse
Affiliation(s)
- Andrew W Simonson
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Todd M Umstead
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA; Pulmonary Immunology and Physiology Laboratory, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Atip Lawanprasert
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Bailey Klein
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sarah Almarzooqi
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - E Scott Halstead
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA; Pulmonary Immunology and Physiology Laboratory, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Scott H Medina
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA; Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA.
| |
Collapse
|
8
|
Baranyai Z, Soria‐Carrera H, Alleva M, Millán‐Placer AC, Lucía A, Martín‐Rapún R, Aínsa JA, la Fuente JM. Nanotechnology‐Based Targeted Drug Delivery: An Emerging Tool to Overcome Tuberculosis. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zsuzsa Baranyai
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Héctor Soria‐Carrera
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - Maria Alleva
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Ana C. Millán‐Placer
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
| | - Ainhoa Lucía
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Rafael Martín‐Rapún
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Departamento de Química Orgánica Facultad de Ciencias Universidad de Zaragoza Zaragoza 50009 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - José A. Aínsa
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús M. la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| |
Collapse
|
9
|
Howard NC, Khader SA. Immunometabolism during Mycobacterium tuberculosis Infection. Trends Microbiol 2020; 28:832-850. [PMID: 32409147 DOI: 10.1016/j.tim.2020.04.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/26/2022]
Abstract
Over a quarter of the world's population is infected with Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Approximately 3.4% of new and 18% of recurrent cases of TB are multidrug-resistant (MDR) or rifampicin-resistant. Recent evidence has shown that certain drug-resistant strains of Mtb modulate host metabolic reprogramming, and therefore immune responses, during infection. However, it remains unclear how widespread these mechanisms are among circulating MDR Mtb strains and what impact drug-resistance-conferring mutations have on immunometabolism during TB. While few studies have directly addressed metabolic reprogramming in the context of drug-resistant Mtb infection, previous literature examining how drug-resistance mutations alter Mtb physiology and differences in the immune response to drug-resistant Mtb provides significant insights into how drug-resistant strains of Mtb differentially impact immunometabolism.
Collapse
Affiliation(s)
- Nicole C Howard
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
10
|
Mayito J, Andia I, Belay M, Jolliffe DA, Kateete DP, Reece ST, Martineau AR. Anatomic and Cellular Niches for Mycobacterium tuberculosis in Latent Tuberculosis Infection. J Infect Dis 2019; 219:685-694. [PMID: 30376080 PMCID: PMC6376907 DOI: 10.1093/infdis/jiy579] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/25/2018] [Indexed: 12/25/2022] Open
Abstract
Latent tuberculosis has been recognized for over a century, but discovery of new niches, where Mycobacterium tuberculosis resides, continues. We evaluated literature on M.tuberculosis locations during latency, highlighting that mesenchymal and hematopoietic stem cells harbor organisms in sensitized asymptomatic individuals.
Collapse
Affiliation(s)
- Jonathan Mayito
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda.,Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Irene Andia
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Mulugeta Belay
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - David A Jolliffe
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - David P Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Stephen T Reece
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Adrian R Martineau
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
11
|
Simonson AW, Lawanprasert A, Goralski TDP, Keiler KC, Medina SH. Bioresponsive peptide-polysaccharide nanogels - A versatile delivery system to augment the utility of bioactive cargo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 17:391-400. [PMID: 30399437 DOI: 10.1016/j.nano.2018.10.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/14/2018] [Accepted: 10/09/2018] [Indexed: 02/02/2023]
Abstract
We report the design, synthesis and efficacy of a new class of gel-like nano-carrier, or 'nanogel', prepared via templated electrostatic assembly of anionic hyaluronic acid (HA) polysaccharides with the cationic peptide amphiphile poly-L-lysine (PLL). Small molecules and proteins present during nanogel assembly become directly encapsulated within the carrier and are precisely released by tuning the nanogel HA:PLL ratio to control particle swelling. Remarkably, nanogels exhibit versatile and complimentary mechanisms of cargo delivery depending on the biologic context. For example, in mammalian cells, nanogels are rapidly internalized and escape the endosome to both deliver membrane-impermeable protein cargo into the cytoplasm and improve chemotherapeutic potency in drug resistant cancer cells. In bacteria, nanogels permeabilize microbial membranes to sensitize bacterial pathogens to the action of a loaded antibiotic. Thus, peptide nanogels represent a versatile, readily scalable and bio-responsive carrier capable of augmenting and enhancing the utility of a broad range of biomolecular cargoes.
Collapse
Affiliation(s)
- Andrew W Simonson
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Atip Lawanprasert
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Tyler D P Goralski
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Kenneth C Keiler
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Scott H Medina
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
12
|
Sasikumar K, Ghosh AR, Dusthackeer A. Antimycobacterial potentials of quercetin and rutin against Mycobacterium tuberculosis H37Rv. 3 Biotech 2018; 8:427. [PMID: 30305996 DOI: 10.1007/s13205-018-1450-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 09/24/2018] [Indexed: 11/30/2022] Open
Abstract
Quercetin and rutin, two flavonoids were examined for antimycobacterial activities against M. tuberculosis H37Rv (ATCC 27294). The quercetin exhibited (99.30 ± 0.268%) in (LRP) assay at 200 µg/ml and 56.21 ± 0.97% inhibition in (BMD) at 50 µg/ml, whereas rutin exhibited (90.40 ± 0.68%) in LRP assay at 200 µg/ml and 56.10 ± 0.67% inhibition in BMD at 50 µg/ml. The minimum inhibitory concentration (MIC) was found to be 6.25 µg ml-1 and 25 µg ml-1 respectively. The current investigation suggests that quercetin has better inhibitory activity than rutin.
Collapse
Affiliation(s)
- Kandasamy Sasikumar
- 1Department of Integrative Biology, School of BioSciences and Technology (SBST), VIT University, Vellore, Tamilnadu 632014 India
| | - Asit Ranjan Ghosh
- 1Department of Integrative Biology, School of BioSciences and Technology (SBST), VIT University, Vellore, Tamilnadu 632014 India
| | - Azger Dusthackeer
- 2Department of Bacteriology, National Institute for Research in Tuberculosis (NIRT-ICMR), Chennai, Tamilnadu 600031 India
| |
Collapse
|
13
|
Luo W, Qu Z, Zhang L, Xie Y, Luo F, Tan Y, Pan Q, Zhang XL. Recombinant BCG::Rv2645 elicits enhanced protective immunity compared to BCG in vivo with induced ISGylation-related genes and Th1 and Th17 responses. Vaccine 2018; 36:2998-3009. [PMID: 29681409 DOI: 10.1016/j.vaccine.2018.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/21/2018] [Accepted: 04/10/2018] [Indexed: 12/27/2022]
Abstract
There is a need to develop protective vaccines against tuberculosis (TB). Recently, we identified an immunodominant T-cell antigen, Rv2645, from the region of deletion 13 (RD13) of M. tuberculosis (M. tb) H37Rv, which is absent in Bacille Calmette-Guérin (BCG). Here, a recombinant BCG expressing Rv2645, namely, BCG::Rv2645, was constructed. Compared to BCG, we found that BCG::Rv2645 improved the antigen presentation capacity of dendritic cells (DCs) and elicited much stronger Th1 and Th17 responses, higher CD44highCD62low effector memory CD4+ T cells (TEM), and fewer T regulated cells (Treg) and regulatory B10 in mice. Importantly, BCG::Rv2645 exhibited enhanced protective efficacy against virulent M. tb H37Rv challenge in both mice and rhesus monkeys, showing less severe pathology and reduced pathogens. Further, transcriptomic analysis and reverse transcription-quantitative real time PCR revealed that the mRNA levels of ISGylation (Isg)-related genes such as interferon-stimulated gene 15 (Isg15), and Th1- and Th17-related genes such as interferon-γ (IFN-γ) and interleukin-17A (IL-17A) were significantly up-regulated in splenocytes and macrophages after stimulation with Rv2645. This study shows that BCG::Rv2645 is a promising TB vaccine candidate with enhanced protective immunity. The enhanced Th1/Th17 immune responses and up-regulation of ISGylation-related genes induced by Rv2645 may be major factors contributing to the protective immunity of BCG::Rv2645.
Collapse
Affiliation(s)
- Wei Luo
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China; Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin 30052, China
| | - Zilu Qu
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Lingyun Zhang
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Yan Xie
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Fengling Luo
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Yang Tan
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Qin Pan
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China.
| |
Collapse
|
14
|
Montanari E, Di Meo C, Oates A, Coviello T, Matricardi P. Pursuing Intracellular Pathogens with Hyaluronan. From a 'Pro-Infection' Polymer to a Biomaterial for 'Trojan Horse' Systems. Molecules 2018; 23:E939. [PMID: 29670009 PMCID: PMC6017551 DOI: 10.3390/molecules23040939] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Hyaluronan (HA) is among the most important bioactive polymers in mammals, playing a key role in a number of biological functions. In the last decades, it has been increasingly studied as a biomaterial for drug delivery systems, thanks to its physico-chemical features and ability to target and enter certain cells. The most important receptor of HA is ‘Cluster of Differentiation 44’ (CD44), a cell surface glycoprotein over-expressed by a number of cancers and heavily involved in HA endocytosis. Moreover, CD44 is highly expressed by keratinocytes, activated macrophages and fibroblasts, all of which can act as ‘reservoirs’ for intracellular pathogens. Interestingly, both CD44 and HA appear to play a key role for the invasion and persistence of such microorganisms within the cells. As such, HA is increasingly recognised as a potential target for nano-carriers development, to pursuit and target intracellular pathogens, acting as a ‘Trojan Horse’. This review describes the biological relationship between HA, CD44 and the entry and survival of a number of pathogens within the cells and the subsequent development of HA-based nano-carriers for enhancing the intracellular activity of antimicrobials.
Collapse
Affiliation(s)
- Elita Montanari
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Chiara Di Meo
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Angela Oates
- School of Healthcare, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK.
| | - Tommasina Coviello
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Pietro Matricardi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
15
|
Hyaluronan Modulation Impacts Staphylococcus aureus Biofilm Infection. Infect Immun 2016; 84:1917-1929. [PMID: 27068096 DOI: 10.1128/iai.01418-15] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/06/2016] [Indexed: 01/06/2023] Open
Abstract
Staphylococcus aureus is a leading cause of chronic biofilm infections. Hyaluronic acid (HA) is a large glycosaminoglycan abundant in mammalian tissues that has been shown to enhance biofilm formation in multiple Gram-positive pathogens. We observed that HA accumulated in an S. aureus biofilm infection using a murine implant-associated infection model and that HA levels increased in a mutant strain lacking hyaluronidase (HysA). S. aureus secretes HysA in order to cleave HA during infection. Through in vitro biofilm studies with HA, the hysA mutant was found to accumulate increased biofilm biomass compared to the wild type, and confocal microscopy showed that HA is incorporated into the biofilm matrix. Exogenous addition of purified HysA enzyme dispersed HA-containing biofilms, while catalytically inactive enzyme had no impact. Additionally, induction of hysA expression prevented biofilm formation and also dispersed an established biofilm in the presence of HA. These observations were corroborated in the implant model, where there was decreased dissemination from an hysA mutant biofilm infection compared to the S. aureus wild type. Histopathology demonstrated that infection with an hysA mutant caused significantly reduced distribution of tissue inflammation compared to wild-type infection. To extend these studies, the impact of HA and S. aureus HysA on biofilm-like aggregates found in joint infections was examined. We found that HA contributes to the formation of synovial fluid aggregates, and HysA can disrupt aggregate formation. Taken together, these studies demonstrate that HA is a relevant component of the S. aureus biofilm matrix and HysA is important for dissemination from a biofilm infection.
Collapse
|
16
|
Naderer T, Heng J, Saunders EC, Kloehn J, Rupasinghe TW, Brown TJ, McConville MJ. Intracellular Survival of Leishmania major Depends on Uptake and Degradation of Extracellular Matrix Glycosaminoglycans by Macrophages. PLoS Pathog 2015; 11:e1005136. [PMID: 26334531 PMCID: PMC4559419 DOI: 10.1371/journal.ppat.1005136] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/10/2015] [Indexed: 02/02/2023] Open
Abstract
Leishmania parasites replicate within the phagolysosome compartment of mammalian macrophages. Although Leishmania depend on sugars as a major carbon source during infections, the nutrient composition of the phagolysosome remains poorly described. To determine the origin of the sugar carbon source in macrophage phagolysosomes, we have generated a N-acetylglucosamine acetyltransferase (GNAT) deficient Leishmania major mutant (∆gnat) that is auxotrophic for the amino sugar, N-acetylglucosamine (GlcNAc). This mutant was unable to grow or survive in ex vivo infected macrophages even when macrophages were cultivated in presence of exogenous GlcNAc. In contrast, the L. major ∆gnat mutant induced normal skin lesions in mice, suggesting that these parasites have access to GlcNAc in tissue macrophages. Intracellular growth of the mutant in ex vivo infected macrophages was restored by supplementation of the macrophage medium with hyaluronan, a GlcNAc-rich extracellular matrix glycosaminoglycan. Hyaluronan is present and constitutively turned-over in Leishmania-induced skin lesions and is efficiently internalized into Leishmania containing phagolysosomes. These findings suggest that the constitutive internalization and degradation of host glycosaminoglycans by macrophages provides Leishmania with essential carbon sources, creating a uniquely favorable niche for these parasites.
Collapse
Affiliation(s)
- Thomas Naderer
- The Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Joanne Heng
- The Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Eleanor C. Saunders
- The Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Joachim Kloehn
- The Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Thusitha W. Rupasinghe
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Tracey J. Brown
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Malcolm J. McConville
- The Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Abstract
Staphylococcus aureus is a Gram-positive pathogen that causes a diverse range of bacterial infections. Invasive S. aureus strains secrete an extensive arsenal of hemolysins, immunomodulators, and exoenzymes to cause disease. Our studies have focused on the secreted enzyme hyaluronidase (HysA), which cleaves the hyaluronic acid polymer at the β-1,4 glycosidic bond. In the study described in this report, we have investigated the regulation and contribution of this enzyme to S. aureus pathogenesis. Using the Nebraska Transposon Mutant Library (NTML), we identified eight insertions that modulate extracellular levels of HysA activity. Insertions in the sigB operon, as well as in genes encoding the global regulators SarA and CodY, significantly increased HysA protein levels and activity. By altering the availability of branched-chain amino acids, we further demonstrated CodY-dependent repression of HysA activity. Additionally, through mutation of the CodY binding box upstream of hysA, the repression of HysA production was lost, suggesting that CodY is a direct repressor of hysA expression. To determine whether HysA is a virulence factor, a ΔhysA mutant of a community-associated methicillin-resistant S. aureus (CA-MRSA) USA300 strain was constructed and found to be attenuated in a neutropenic, murine model of pulmonary infection. Mice infected with this mutant strain exhibited a 4-log-unit reduction in bacterial burden in their lungs, as well as reduced lung pathology and increased levels of pulmonary hyaluronic acid, compared to mice infected with the wild-type, parent strain. Taken together, these results indicate that S. aureus hyaluronidase is a CodY-regulated virulence factor.
Collapse
|
18
|
Aptamer against mannose-capped lipoarabinomannan inhibits virulent Mycobacterium tuberculosis infection in mice and rhesus monkeys. Mol Ther 2014; 22:940-51. [PMID: 24572295 DOI: 10.1038/mt.2014.31] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 02/20/2014] [Indexed: 12/23/2022] Open
Abstract
The major surface lipoglycan of Mycobacterium tuberculosis (M. tb), mannose-capped lipoarabinomannan (ManLAM), is an immunosuppressive epitope of M. tb. We used systematic evolution of ligands by exponential enrichment (SELEX) to generate an aptamer (ZXL1) that specifically bound to ManLAM from the virulent M. tb strain H37Rv. Aptamer ZXL1 had the highest binding affinity, with an equilibrium dissociation constant (Kd) of 436.3 ± 37.84 nmol/l, and competed with the mannose receptor for binding to ManLAM and M. tb H37Rv. ZXL1 significantly inhibited the ManLAM-induced immunosuppression of CD11c(+) dendritic cells (DCs) and enhanced the M. tb antigen-presenting activity of DCs for naive CD4(+) Th1 cell activation. More importantly, we demonstrated that injection of aptamer ZXL1 significantly reduced the progression of M. tb H37Rv infections and bacterial loads in lungs of mice and rhesus monkeys. These results suggest that the aptamer ZXL1 is a new potential antimycobacterial agent and tuberculosis vaccine immune adjuvant.
Collapse
|
19
|
MacDonald LC, Berger BW. A polysaccharide lyase from Stenotrophomonas maltophilia with a unique, pH-regulated substrate specificity. J Biol Chem 2013; 289:312-25. [PMID: 24257754 DOI: 10.1074/jbc.m113.489195] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Polysaccharide lyases (PLs) catalyze the depolymerization of anionic polysaccharides via a β-elimination mechanism. PLs also play important roles in microbial pathogenesis, participating in bacterial invasion and toxin spread into the host tissue via degradation of the host extracellular matrix, or in microbial biofilm formation often associated with enhanced drug resistance. Stenotrophomonas maltophilia is a Gram-negative bacterium that is among the emerging multidrug-resistant organisms associated with chronic lung infections as well as with cystic fibrosis patients. A putative alginate lyase (Smlt1473) from S. maltophilia was heterologously expressed in Escherichia coli, purified in a one-step fashion via affinity chromatography, and activity as well as specificity determined for a range of polysaccharides. Interestingly, Smlt1473 catalyzed the degradation of not only alginate, but poly-β-D-glucuronic acid and hyaluronic acid as well. Furthermore, the pH optimum for enzymatic activity is substrate-dependent, with optimal hyaluronic acid degradation at pH 5, poly-β-D-glucuronic acid degradation at pH 7, and alginate degradation at pH 9. Analysis of the degradation products revealed that each substrate was cleaved endolytically into oligomers comprised predominantly of even numbers of sugar groups, with lower accumulation of trimers and pentamers. Collectively, these results imply that Smlt1473 is a multifunctional PL that exhibits broad substrate specificity, but utilizes pH as a mechanism to achieve selectivity.
Collapse
|
20
|
Yuan X, Chen L, Deng X, Cao J, Yu S, Quankai W, Pang H, Liu S. Characterization of Rv0394c gene encoding hyaluronidase and chondrosulfatase from Mycobacterium tuberculosis. Tuberculosis (Edinb) 2013; 93:296-300. [PMID: 23465892 DOI: 10.1016/j.tube.2013.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/16/2013] [Accepted: 01/27/2013] [Indexed: 10/27/2022]
Abstract
It has been demonstrated that Mycobacterium tuberculosis can utilize hyaluronan (HA) as an alternative carbon resource; however, the gene responsible for HA utilization has not been identified. We overexpressed the soluble product of the Rv0394c gene from M. tuberculosis H37Rv in Escherichia coli and purified it using affinity chromatography and anion exchange chromatography. The hyaluronidase and chondrosulfatase activities of the purified recombinant protein Rv0394c (rRv0394c) were detected and quantitatively measured. Analysis of nucleotide and derived amino acid sequences of the Rv0394c gene revealed that homologs of this gene were conserved in pathogenic mycobacteria, but absent in non-pathogenic mycobacteria. In the current study, we provide novel identification and characterization of a gene encoding hyaluronidase and chondrosulfatase in M. tuberculosis.
Collapse
Affiliation(s)
- Xianglian Yuan
- Division of Bacterial Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 427 Maduan Street , Nangang Dist., Harbin 15000, PR China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Bordbar A, Mo ML, Nakayasu ES, Schrimpe-Rutledge AC, Kim YM, Metz TO, Jones MB, Frank BC, Smith RD, Peterson SN, Hyduke DR, Adkins JN, Palsson BO. Model-driven multi-omic data analysis elucidates metabolic immunomodulators of macrophage activation. Mol Syst Biol 2012; 8:558. [PMID: 22735334 PMCID: PMC3397418 DOI: 10.1038/msb.2012.21] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 05/09/2012] [Indexed: 12/11/2022] Open
Abstract
Macrophages are central players in immune response, manifesting divergent phenotypes to control inflammation and innate immunity through release of cytokines and other signaling factors. Recently, the focus on metabolism has been reemphasized as critical signaling and regulatory pathways of human pathophysiology, ranging from cancer to aging, often converge on metabolic responses. Here, we used genome-scale modeling and multi-omics (transcriptomics, proteomics, and metabolomics) analysis to assess metabolic features that are critical for macrophage activation. We constructed a genome-scale metabolic network for the RAW 264.7 cell line to determine metabolic modulators of activation. Metabolites well-known to be associated with immunoactivation (glucose and arginine) and immunosuppression (tryptophan and vitamin D3) were among the most critical effectors. Intracellular metabolic mechanisms were assessed, identifying a suppressive role for de-novo nucleotide synthesis. Finally, underlying metabolic mechanisms of macrophage activation are identified by analyzing multi-omic data obtained from LPS-stimulated RAW cells in the context of our flux-based predictions. Our study demonstrates metabolism's role in regulating activation may be greater than previously anticipated and elucidates underlying connections between activation and metabolic effectors.
Collapse
Affiliation(s)
- Aarash Bordbar
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Monica L Mo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | | | | | - Young-Mo Kim
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Thomas O Metz
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Bryan C Frank
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | | | | - Daniel R Hyduke
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | | | - Bernhard O Palsson
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Matsumoto S. [Analysis of molecular mechanisms of the virulence and growth coordination of Mycobacterium tuberculosis]. Nihon Saikingaku Zasshi 2011; 66:531-537. [PMID: 22214749 DOI: 10.3412/jsb.66.531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Affiliation(s)
- Sohkichi Matsumoto
- Department of Bacteriology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| |
Collapse
|
23
|
Lennon FE, Singleton PA. Role of hyaluronan and hyaluronan-binding proteins in lung pathobiology. Am J Physiol Lung Cell Mol Physiol 2011; 301:L137-47. [PMID: 21571904 DOI: 10.1152/ajplung.00071.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hyaluronan (HA) has diverse functions in normal lung homeostasis and pulmonary disease. HA constitutes the major glycosaminoglycan in lung tissue, with HA degradation products, produced by hyaluronidase enzymes and reactive oxygen species, being implicated in several lung diseases, including acute lung injury, asthma, chronic obstructive pulmonary disease, and pulmonary hypertension. The differential activities of HA and its degradation products are due, in part, to regulation of multiple HA-binding proteins, including cluster of differentiation 44 (CD44), Toll-like receptor 4 (TLR4), HA-binding protein 2 (HABP2), and receptor for HA-mediated motility (RHAMM). Recent research indicates that exogenous administration of high-molecular-weight HA can serve as a novel therapeutic intervention for lung diseases, including lipopolysaccharide (LPS)-induced acute lung injury, sepsis/ventilator-induced lung injury, and airway hyperreactivity. This review focuses on the regulatory role of HA and HA-binding proteins in lung pathology and discusses the capacity of HA to augment and inhibit various lung diseases.
Collapse
Affiliation(s)
- Frances E Lennon
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
24
|
Insight into human alveolar macrophage and M. tuberculosis interactions via metabolic reconstructions. Mol Syst Biol 2011; 6:422. [PMID: 20959820 PMCID: PMC2990636 DOI: 10.1038/msb.2010.68] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 07/30/2010] [Indexed: 02/06/2023] Open
Abstract
A human alveolar macrophage genome-scale metabolic reconstruction was reconstructed from tailoring a global human metabolic network, Recon 1, by using computational algorithms and manual curation. A genome-scale host–pathogen network of the human alveolar macrophage and Mycobacterium tuberculosis is presented. This involved integrating two genome-scale network reconstructions. The reaction activity and gene essentiality predictions of the host–pathogen model represent a more accurate depiction of infection. Integration of high-throughput data into a host-pathogen model followed by systems analysis was performed in order to elucidate major metabolic differences under different types of M. tuberculosis infection.
Mycobacterium tuberculosis (M. tb) is an insidious and highly persistent pathogen that affects one-third of the world's population (WHO, 2009). Metabolism is foundational to M. tb's infection ability and the ensuing host–pathogen interactions. In addition, M. tb has a heterogeneous clinical presentation and can infect virtually every tissue. Depending on the location of the infection, different metabolic pathways are active and inactive in both the host and pathogen cells. In this study, we sought to model the host–pathogen interactions of the human alveolar macrophage and M. tb as well as detail the metabolic differences in specific infection types using genome-scale metabolic reconstructions (Figure 4A). Genome-scale metabolic reconstructions are knowledge bases of all known metabolic reactions of a given organism. Reconstructions have been shown to elucidate the mechanistic genotype-to-phenotype relationship through the integration of high-throughput and physiological data (Oberhardt et al, 2009). Genome-scale reconstructions are converted into mathematical models under the constraints-based reconstruction and analysis (COBRA) platform (Becker et al, 2007). COBRA models use network stoichiometry and steady-state mass balances to define a solution space of potential flux states that a network can take. Thus, the COBRA approach does not require kinetic parameters. Recently, the global human metabolic network, Recon 1, has been reconstructed (Duarte et al, 2007). To understand the metabolic host–pathogen integrations of M. tb with its human host, we first tailored the global human metabolic network into a cell-specific metabolic reconstruction of the human alveolar macrophage. This was carried out using established computational algorithms (Becker and Palsson, 2008; Shlomi et al, 2008) and manual curation to confirm the included and excluded reactions. The human alveolar macrophage reconstruction, iAB-AMØ-1410, accounts for 1410 genes, 3012 intracellular reactions, and 2572 metabolites (Figure 4C). iAB-AMØ-1410 was able to accurately predict maximum ATP and NO production rates obtained from experimental data (Griscavage et al, 1993; Newsholme et al, 1999). The second step to studying host–pathogen interactions was integration of the human alveolar macrophage reconstruction with an existing genome-scale metabolic model of M. tb, iNJ661 (Jamshidi and Palsson, 2007). Interfacial constraints were set to create a phagosomal environment that was hypoxic, nitrosative, rich in fatty acids, and poor in carbohydrates. From the onset, it was apparent that some oxygen (<15% of in vitro uptake) was required for proper simulations. In addition, algorithmic tailoring of the M. tb biomass objective function was performed to better represent an infectious state. The integrated host–pathogen metabolic reconstruction was dubbed iAB-AMØ-1410-Mt-661. Analysis of the integrated host–pathogen metabolic reconstruction resulted in three main findings. First, by setting interfacial constraints and tailoring the biomass objective function, the solution space better represents an infectious state. Without adding artificial constraints to the host portion of the integrated model, the iAB-AMØ-1410 solution space is greatly reduced (Figure 4B). Macrophage glycolysis and nitric oxide production are up-regulated and macrophage ATP production, nucleotide synthesis, and amino-acid metabolism are suppressed. In addition, M. tb glycolysis is suppressed and isocitrate lyase is up-regulated for generation of acetyl-CoA. Fatty acid oxidation pathways and production of mycolic acids are increased, while production of nucleotides, peptidoglycans, and phenolic glycolipids are reduced. The modified solution space of the alveolar macrophage and M. tb better represents the infectious state. Second, the host-pathogen model more accurately predicts M. tb gene deletion tests than the current in vitro model, iNJ661. The host-pathogen model predicted 11 essential genes and 37 unessential genes differently than iNJ661. A total of 22 of the differentially predicted genes have been experimentally characterized (Sassetti and Rubin, 2003; Sohaskey, 2008). The host-pathogen model correctly predicted 18 of the 22 genes. Thus, iAB-AMØ-1410-Mt-661 is a more accurate platform for studying infectious states of M. tb. Finally, we sought to determine metabolic differences in both the macrophage and M. tb between three different types of infection: latent, pulmonary, and meningeal. Transcription profiling data of the macrophage for the three infections (Thuong et al, 2008) were integrated in the context of the host–pathogen network to elucidate the reaction activity of the three infections. There was wide heterogeneity in the three infection states; some of these differences are highlighted. Macrophage hyaluronan synthase and export were only active in the pulmonary infection. This is potentially interesting from a pharmaceutical viewpoint as hyaluronan has been implicated as a potential carbon source for extracellular M. tb (Hirayama et al, 2009). In addition, we detected metabolic activity differences in M. tb pathways that have been previously discussed as potential drug targets (Eoh et al, 2007; Boshoff et al, 2008). Polyprenyl metabolic reactions were only active in the latent state infection, while de novo synthesis of nicotinamide cofactors was only active in latent and meningeal M. tb infections. Host-pathogen modeling represents a novel approach for studying metabolic interactions during infection. iAB-AMØ-1410-Mt-661 is a more accurate platform for understanding the biology and pathophysiology of M. tb infection. Most importantly, genome-scale metabolic reconstructions can act as scaffolds for integrating high-throughput data. Particularly, in this study we were able to discern reaction activity differences between different infection types. Metabolic coupling of Mycobacterium tuberculosis to its host is foundational to its pathogenesis. Computational genome-scale metabolic models have shown utility in integrating -omic as well as physiologic data for systemic, mechanistic analysis of metabolism. To date, integrative analysis of host–pathogen interactions using in silico mass-balanced, genome-scale models has not been performed. We, therefore, constructed a cell-specific alveolar macrophage model, iAB-AMØ-1410, from the global human metabolic reconstruction, Recon 1. The model successfully predicted experimentally verified ATP and nitric oxide production rates in macrophages. This model was then integrated with an M. tuberculosis H37Rv model, iNJ661, to build an integrated host–pathogen genome-scale reconstruction, iAB-AMØ-1410-Mt-661. The integrated host–pathogen network enables simulation of the metabolic changes during infection. The resulting reaction activity and gene essentiality targets of the integrated model represent an altered infectious state. High-throughput data from infected macrophages were mapped onto the host–pathogen network and were able to describe three distinct pathological states. Integrated host–pathogen reconstructions thus form a foundation upon which understanding the biology and pathophysiology of infections can be developed.
Collapse
|
25
|
Dhiman R, Bandaru A, Barnes PF, Saha S, Tvinnereim A, Nayak RC, Paidipally P, Valluri VL, Rao LVM, Vankayalapati R. c-Maf-dependent growth of Mycobacterium tuberculosis in a CD14(hi) subpopulation of monocyte-derived macrophages. THE JOURNAL OF IMMUNOLOGY 2011; 186:1638-45. [PMID: 21209279 DOI: 10.4049/jimmunol.1003146] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Macrophages are a major component of the innate immune response, comprising the first line of defense against various intracellular pathogens, including Mycobacterium tuberculosis. In this report, we studied the factors that regulate growth of M. tuberculosis H37Rv in subpopulations of human monocyte-derived macrophages (MDMs). In healthy donors, M. tuberculosis H37Rv grew 5.6-fold more rapidly in CD14(hi) MDMs compared with that in CD14(lo)CD16(+) MDMs. Compared with CD14(lo)CD16(+) cells, M. tuberculosis H37Rv-stimulated CD14(hi) monocytes produced more IL-10 and had increased mRNA expression for c-Maf, a transcription factor that upregulates IL-10 gene expression. c-Maf small interfering RNA (siRNA) inhibited IL-10 production and growth of M. tuberculosis in CD14(hi) cells. Compared with CD14(lo)CD16(+) monocytes, M. tuberculosis H37Rv-stimulated CD14(hi) cells had increased expression of 22 genes whose promoters contained a c-Maf binding site, including hyaluronan synthase 1 (HAS1). c-Maf siRNA inhibited HAS1 expression in M. tuberculosis-stimulated CD14(hi) monocytes, and HAS1 siRNA inhibited growth of M. tuberculosis in CD14(hi) MDMs. M. tuberculosis H37Rv upregulated expression of HAS1 protein and its product, hyaluronan, in CD14(hi) MDMs. We conclude that M. tuberculosis grows more rapidly in CD14(hi) than in CD14(lo)CD16(+) MDMs because CD14(hi) cells have increased expression of c-Maf, which increases production of two key factors (hyaluronan and IL-10) that promote growth of M. tuberculosis.
Collapse
Affiliation(s)
- Rohan Dhiman
- Center for Pulmonary and Infectious Disease Control, University of Texas Health Science Center, Tyler, TX 75708, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mycobacterium tuberculosis eis regulates autophagy, inflammation, and cell death through redox-dependent signaling. PLoS Pathog 2010; 6:e1001230. [PMID: 21187903 PMCID: PMC3002989 DOI: 10.1371/journal.ppat.1001230] [Citation(s) in RCA: 254] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 11/15/2010] [Indexed: 11/19/2022] Open
Abstract
The “enhanced intracellular survival” (eis) gene of Mycobacterium tuberculosis (Mtb) is involved in the intracellular survival of M. smegmatis. However, its exact effects on host cell function remain elusive. We herein report that Mtb Eis plays essential roles in modulating macrophage autophagy, inflammatory responses, and cell death via a reactive oxygen species (ROS)-dependent pathway. Macrophages infected with an Mtb eis-deletion mutant H37Rv (Mtb-Δeis) displayed markedly increased accumulation of massive autophagic vacuoles and formation of autophagosomes in vitro and in vivo. Infection of macrophages with Mtb-Δeis increased the production of tumor necrosis factor-α and interleukin-6 over the levels produced by infection with wild-type or complemented strains. Elevated ROS generation in macrophages infected with Mtb-Δeis (for which NADPH oxidase and mitochondria were largely responsible) rendered the cells highly sensitive to autophagy activation and cytokine production. Despite considerable activation of autophagy and proinflammatory responses, macrophages infected with Mtb-Δeis underwent caspase-independent cell death. This cell death was significantly inhibited by blockade of autophagy and c-Jun N-terminal kinase-ROS signaling, suggesting that excessive autophagy and oxidative stress are detrimental to cell survival. Finally, artificial over-expression of Eis or pretreatment with recombinant Eis abrogated production of both ROS and proinflammatory cytokines, which depends on the N-acetyltransferase domain of the Eis protein. Collectively, these data indicate that Mtb Eis suppresses host innate immune defenses by modulating autophagy, inflammation, and cell death in a redox-dependent manner. Tuberculosis is a global health problem: at least one-third of the world's population is infected with Mycobacterium tuberculosis (Mtb). Mtb is a successful pathogen that enhances its own intracellular survival by arresting phagolysosomal fusion. Recently, autophagy has emerged as a host defense strategy against Mtb infection, through stimulation of the fusion of phagosomes and lysosomes. However, excessive and uncontrolled autophagic activity can be detrimental to host cells and can result in their death. The Mtb “enhanced intracellular survival” (eis) gene has been implicated in the intracellular survival of M. smegmatis. However, its exact role and how it regulates host innate immune responses have not been fully explained. Here, we provide evidence that Eis suppresses macrophage autophagy, inflammation, and cell death through the inhibition of reactive oxygen species (ROS) generation. Although it has previously been demonstrated that autophagy is a key host defense response to mycobacterial infections, our data indicate that excessive autophagy, and the resulting cell death, do not significantly affect host defense responses to mycobacteria. Additionally, our data reveal that Eis's ability to regulate ROS generation and proinflammatory responses depends on its N-acetyltransferase domain. These results underscore a previously unrecognized role of Eis in modulating host inflammatory responses, oxidative stress, and cell survival/death during mycobacterial infection.
Collapse
|