1
|
Li J, Foged C. Evaluating the breadth of nucleic acid-based payloads delivered in lipid nanoparticles to establish fundamental differences in development. Expert Opin Drug Deliv 2024; 21:1441-1461. [PMID: 39387233 DOI: 10.1080/17425247.2024.2409142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Nucleic acid (NA)-based therapeutics have shown great potential for downregulating or augmenting gene expression, and for promising applications, e.g., protein-replacement therapy and vaccination, a comprehensive understanding of the requirements for their targeted delivery to specific tissues or cells is needed. AREAS COVERED In this review, we discuss clinical applications of four representative types of NA-based therapeutics, i.e. antisense oligonucleotides, small interfering RNA, messenger RNA, and circular RNA, with a focus on the lipid nanoparticle (LNP) technology used for intracellular delivery. The in vivo fate of LNPs is discussed to improve the understanding of trafficking of nanomedicines at the systemic and cellular levels. In addition, NA-based vaccines are discussed, focusing on targeting antigen-presenting cells and immune activation. EXPERT OPINION Optimization of delivery systems for NA-based therapeutics is mainly focused on the standard requirements of prolonged systemic circulation and enhancing endosomal escape. Depending on the final destination in specific target tissues or cells, strategies should be adjusted to achieve the desired biodistribution of NA-based payloads. More studies relating to the pharmacokinetics of both cargo and carrier are encouraged, because their in vivo fates may differ, considering the possibility of premature cargo release before reaching the target.
Collapse
Affiliation(s)
- Jinjin Li
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| |
Collapse
|
2
|
Chang SR, Chou CH, Tu HF, Liu CJ, Chang KW, Lin SC. The expression of immune co-stimulators as a prognostic predictor of head and neck squamous cell carcinomas and oral squamous cell carcinomas. J Dent Sci 2024; 19:1380-1388. [PMID: 39035328 PMCID: PMC11259670 DOI: 10.1016/j.jds.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose T cells require second immune checkpoint molecules for activation and immune memory after antigen presentation. We found that inducible co-stimulator (ICOS) has been a favorable prognostic factor amongst B7 immune checkpoint co-stimulators (ICSs) families in head and neck squamous cell carcinoma (HNSCC) and oral SCC (OSCC). Materials and methods This study analyzed the expression of non-B7 tumor necrosis factor (TNF) superfamily ICSs in the Cancer Genome Atlas (TCGA) HNSCC cohort, our OSCC cohort, and TCGA pan-cancer datasets. The correlation in expression, prognosis, and immune status was assessed. Results The higher expression of CD27, CD30, CD40L, death domain 3 (DR3), and OX40, presumably on the T cell surface, defined better overall survival of HNSCC patients. Besides, CD27, CD30, CD40L, and OX40 were highly correlated with ICOS expression in tumors. CD27, CD40L, and DR3 expression are higher in HPV+ HNSCC tumors than in HPV- tumors. The combined expression level of CD27/OX40 or CD27/CD40L/OX40 enables the potent survival prediction of small, less nodal involvement, early stage, and HPV + tumor subsets. Tumors expressing high CD27, CD30, CD40L, ICOS, and OX40 exhibited enhanced immune cell infiltration. The high correlation in the expression of these ICSs was also noted in the vast majority of tumor types in TCGA datasets. Conclusion The findings of this study not only confirm the potential of the concordant stimulation of CD27, CD30, CD40L, ICOS, and OX40 as a crucial strategy in cancer immunotherapy but also inspire further exploration into the field, highlighting the promising future of cancer treatment.
Collapse
Affiliation(s)
- Shi-Rou Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Hsien Chou
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsi-Feng Tu
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, Taipei Mackay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
3
|
Bhatnagar A, Chopra U, Raja S, Das KD, Mahalingam S, Chakravortty D, Srinivasula SM. TLR-mediated aggresome-like induced structures comprise antimicrobial peptides and attenuate intracellular bacterial survival. Mol Biol Cell 2024; 35:ar34. [PMID: 38170582 PMCID: PMC10916861 DOI: 10.1091/mbc.e23-09-0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Immune cells employ diverse mechanisms for host defense. Macrophages, in response to TLR activation, assemble aggresome-like induced structures (ALIS). Our group has shown TLR4-signaling transcriptionally upregulates p62/sequestome1, which assembles ALIS. We have demonstrated that TLR4-mediated autophagy is, in fact, selective-autophagy of ALIS. We hypothesize that TLR-mediated autophagy and ALIS contribute to host-defense. Here we show that ALIS are assembled in macrophages upon exposure to different bacteria. These structures are associated with pathogen-containing phagosomes. Importantly, we present evidence of increased bacterial burden, where ALIS assembly is prevented with p62-specific siRNA. We have employed 3D-super-resolution structured illumination microscopy (3D-SR-SIM) and mass-spectrometric (MS) analyses to gain insight into the assembly of ALIS. Ultra-structural analyses of known constituents of ALIS (p62, ubiquitin, LC3) reveal that ALIS are organized structures with distinct patterns of alignment. Furthermore, MS-analyses of ALIS identified, among others, several proteins of known antimicrobial properties. We have validated MS data by testing the association of some of these molecules (Bst2, IFITM2, IFITM3) with ALIS and the phagocytosed-bacteria. We surmise that AMPs enrichment in ALIS leads to their delivery to bacteria-containing phagosomes and restricts the bacteria. Our findings in this paper support hitherto unknown functions of ALIS in host-defense.
Collapse
Affiliation(s)
- Anushree Bhatnagar
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Umesh Chopra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sebastian Raja
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - S. Mahalingam
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Dipshikha Chakravortty
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Srinivasa Murty Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| |
Collapse
|
4
|
Luz IS, Takaya R, Ribeiro DG, Castro MS, Fontes W. Proteomics: Unraveling the Cross Talk Between Innate Immunity and Disease Pathophysiology, Diagnostics, and Treatment Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:221-242. [PMID: 38409424 DOI: 10.1007/978-3-031-50624-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Inflammation is crucial in diseases, and proteins play a key role in the interplay between innate immunity and pathology. This review explores how proteomics helps understanding this relationship, focusing on diagnosis and treatment. We explore the dynamic innate response and the significance of proteomic techniques in deciphering the complex network of proteins involved in prevalent diseases, including infections, cancer, autoimmune and neurodegenerative disorders. Proteomics identifies key proteins in host-pathogen interactions, shedding light on infection mechanisms and inflammation. These discoveries hold promise for diagnostic tools, therapies, and vaccines. In cancer research, proteomics reveals innate signatures associated with tumor development, immune evasion, and therapeutic response. Additionally, proteomic analysis has unveiled autoantigens and dysregulation of the innate immune system in autoimmunity, offering opportunities for early diagnosis, disease monitoring, and new therapeutic targets. Moreover, proteomic analysis has identified altered protein expression patterns in neurodegenerative diseases like Alzheimer's and Parkinson's, providing insights into potential therapeutic strategies. Proteomics of the innate immune system provides a comprehensive understanding of disease mechanisms, identifies biomarkers, and enables effective interventions in various diseases. Despite still in its early stages, this approach holds great promise to revolutionize innate immunity research and significantly improve patient outcomes across a wide range of diseases.
Collapse
Affiliation(s)
- Isabelle Souza Luz
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Raquel Takaya
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Daiane Gonzaga Ribeiro
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Mariana S Castro
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil.
| |
Collapse
|
5
|
Ren W, Zhao L, Sun Y, Wang X, Shi X. HMGB1 and Toll-like receptors: potential therapeutic targets in autoimmune diseases. Mol Med 2023; 29:117. [PMID: 37667233 PMCID: PMC10478470 DOI: 10.1186/s10020-023-00717-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023] Open
Abstract
HMGB1, a nucleoprotein, is expressed in almost all eukaryotic cells. During cell activation and cell death, HMGB1 can function as an alarm protein (alarmin) or damage-associated molecular pattern (DAMP) and mediate early inflammatory and immune response when it is translocated to the extracellular space. The binding of extracellular HMGB1 to Toll-like receptors (TLRs), such as TLR2 and TLR4 transforms HMGB1 into a pro-inflammatory cytokine, contributing to the occurrence and development of autoimmune diseases. TLRs, which are members of a family of pattern recognition receptors, can bind to endogenous DAMPs and activate the innate immune response. Additionally, TLRs are key signaling molecules mediating the immune response and play a critical role in the host defense against pathogens and the maintenance of immune balance. HMGB1 and TLRs are reported to be upregulated in several autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, type 1 diabetes mellitus, and autoimmune thyroid disease. The expression levels of HMGB1 and some TLRs are upregulated in tissues of patients with autoimmune diseases and animal models of autoimmune diseases. The suppression of HMGB1 and TLRs inhibits the progression of inflammation in animal models. Thus, HMGB1 and TLRs are indispensable biomarkers and important therapeutic targets for autoimmune diseases. This review provides comprehensive strategies for treating or preventing autoimmune diseases discovered in recent years.
Collapse
Affiliation(s)
- Wenxuan Ren
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Lei Zhao
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Ying Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Xichang Wang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Xiaoguang Shi
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
6
|
Tsai CN, Massicotte MA, MacNair CR, Perry JN, Brown ED, Coombes BK. Screening under infection-relevant conditions reveals chemical sensitivity in multidrug resistant invasive non-typhoidal Salmonella (iNTS). RSC Chem Biol 2023; 4:600-612. [PMID: 37547457 PMCID: PMC10398353 DOI: 10.1039/d3cb00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Bloodstream infections caused by invasive, non-typhoidal Salmonella (iNTS) are a major global health concern, particularly in Africa where the pathogenic variant of Salmonella Typhimurium sequence type (ST) 313 is dominant. Unlike S. Typhimurium strains that cause gastroenteritis, iNTS strains cause bloodstream infections and are resistant to multiple first-line antibiotics, thus limiting current treatment options. Here, we developed and implemented multiple small molecule screens under physiological, infection-relevant conditions to reveal chemical sensitivities in ST313 and to identify host-directed therapeutics as entry points to drug discovery to combat the clinical burden of iNTS. Screening ST313 iNTS under host-mimicking growth conditions identified 92 compounds with antimicrobial activity despite inherent multidrug resistance. We characterized the antimicrobial activity of the nucleoside analog 3'-azido-3'-deoxythymidine as an exemplary compound from this screen, which depended on bacterial thymidine kinase activity for antimicrobial activity. In a companion macrophage-based screening platform designed to enrich for host-directed therapeutics, we identified three compounds (amodiaquine, berbamine, and indatraline) as actives that required the presence of host cells for antibacterial activity. These three compounds had antimicrobial activity only in the presence of host cells that significantly inhibited intracellular ST313 iNTS replication in macrophages. This work provides evidence that despite high invasiveness and multidrug resistance, ST313 iNTS remains susceptible to unconventional drug discovery approaches.
Collapse
Affiliation(s)
- Caressa N Tsai
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
- Michael G. DeGroote Institute for Infectious Disease Research Hamilton ON Canada
| | - Marie-Ange Massicotte
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
- Michael G. DeGroote Institute for Infectious Disease Research Hamilton ON Canada
| | - Craig R MacNair
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
- Michael G. DeGroote Institute for Infectious Disease Research Hamilton ON Canada
| | - Jordyn N Perry
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
| | - Eric D Brown
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
- Michael G. DeGroote Institute for Infectious Disease Research Hamilton ON Canada
| | - Brian K Coombes
- Department of Biochemistry & Biomedical Sciences, McMaster University Hamilton ON L8S 4L8 Canada
- Michael G. DeGroote Institute for Infectious Disease Research Hamilton ON Canada
- Farncombe Family Digestive Health Research Institute Hamilton ON Canada
| |
Collapse
|
7
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
8
|
Ohto U. Activation and regulation mechanisms of NOD-like receptors based on structural biology. Front Immunol 2022; 13:953530. [PMID: 36189327 PMCID: PMC9520476 DOI: 10.3389/fimmu.2022.953530] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immunity is a primary defense system against microbial infections. Innate immune pattern recognition receptors (PRRs) play pivotal roles in detection of invading pathogens. When pathogens, such as bacteria and viruses, invade our bodies, their components are recognized by PRRs as pathogen-associated molecular patterns (PAMPs), activating the innate immune system. Cellular components such as DNA and RNA, acting as damage-associated molecular patterns (DAMPs), also activate innate immunity through PRRs under certain conditions. Activation of PRRs triggers inflammatory responses, interferon-mediated antiviral responses, and the activation of acquired immunity. Research on innate immune receptors is progressing rapidly. A variety of these receptors has been identified, and their regulatory mechanisms have been elucidated. Nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) constitute a major family of intracellular PRRs and are involved in not only combating pathogen invasion but also maintaining normal homeostasis. Some NLRs are known to form multi-protein complexes called inflammasomes, a process that ultimately leads to the production of inflammatory cytokines and induces pyroptosis through the proteolytic cascade. The aberrant activation of NLRs has been found to be associated with autoimmune diseases. Therefore, NLRs are considered targets for drug discovery, such as for antiviral drugs, immunostimulants, antiallergic drugs, and autoimmune disease drugs. This review summarizes our recent understanding of the activation and regulation mechanisms of NLRs, with a particular focus on their structural biology. These include NOD2, neuronal apoptosis inhibitory protein (NAIP)/NLRC4, NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLRP6, and NLRP9. NLRs are involved in a variety of diseases, and their detailed activation mechanisms based on structural biology can aid in developing therapeutic agents in the future.
Collapse
|
9
|
Abu Owida H, Al-Nabulsi JI, Alnaimat F, Al Sharah A, Al-Ayyad M, Turab NM, Abdullah M. Advancement of Nanofibrous Mats and Common Useful Drug Delivery Applications. Adv Pharmacol Pharm Sci 2022; 2022:9073837. [PMID: 35492808 PMCID: PMC9042622 DOI: 10.1155/2022/9073837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 11/18/2022] Open
Abstract
Electrospinning enables simple and cost-effective production of polymer nanofibers from different polymer materials. Drug delivery systems are capable of achieving maximum drug treatment benefits by significantly reducing adverse complications. Electrospun nanofibers have recently attracted considerable attention owing to their distinctive properties, including flexibility and biocompatibility. The implementation of functional constituents within nanostructure fibers blends is an effective technique for the administration of a variety of drugs in animal research, broadening the nanofiber capability and reliability. The nanofibrous mesh and its various application purposes are discussed in terms of a summary of recent research, emphasizing the ease of streaming and a large number of combinations of this approach, which could lead to a breakthrough in targeted therapy.
Collapse
Affiliation(s)
- Hamza Abu Owida
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Jamal I. Al-Nabulsi
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Feras Alnaimat
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ashraf Al Sharah
- Computer Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Muhammad Al-Ayyad
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Nidal M. Turab
- Department of Networks and Information Security, Faculty of Information Technology, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Mustafa Abdullah
- Civil Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| |
Collapse
|
10
|
Guryanova SV, Gigani OB, Gudima GO, Kataeva AM, Kolesnikova NV. Dual Effect of Low-Molecular-Weight Bioregulators of Bacterial Origin in Experimental Model of Asthma. Life (Basel) 2022; 12:192. [PMID: 35207480 PMCID: PMC8879587 DOI: 10.3390/life12020192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Asthma is one of the most common noncommunicable diseases, affecting over 200 million people. A large number of drugs control asthma attacks, but there is no effective therapy. Identification of reasons for asthma and preventing this disease is a relevant task. The influence of bacterial components is necessary for the normal development of the immune system and the formation of an adequate immune response to antigens. In the absence of microorganisms or their insufficient exposure, the prerequisites are formed for excessive reactivity to harmless antigens. In the present study, we analyzed cellular and humoral factors in a standard mouse model of OVA-induced asthma modified by 5-fold intraperitoneal injection of bacterial cell wall fragments of glucosaminylmuramyl dipeptide (GMDP) 5 μg/animal or 1 μg lipopolysaccharide (LPS) per animal for 5 days before sensitization by ovalbumin (OVA). Preliminary administration of LPS or GMDP to animals significantly reduced goblet cells as well as the number of neutrophils, lymphocytes, and eosinophils in bronchoalveolar lavage, wherein GMDP corrected neutrophilia to a 2-fold degree, and LPS reduced the severity of eosinophilia by 1.9 times. With OVA administration of GMDP or LPS at the sensitization stage, an increase in the total number of bronchoalveolar lavage cells due to neutrophils, macrophages, lymphocytes, and eosinophils in relation to the group with asthma without GMDP or LPS was observed. The administration of GMDP or LPS to normal mice without asthma for 5 days had no statistically significant effect on the change in the number and population composition of cells in bronchoalveolar lavage in comparison with the control group receiving PBS. As a result of a study in a mouse model of asthma, a dual effect of LPS and GMDP was established: the introduction of LPS or GMDP before sensitization reduces neutrophilia and eosinophilia, while the introduction of LPS or GMDP together with an allergen significantly increases neutrophilia and eosinophilia. The study of the immunoglobulin status shows that in normal-asthma mice, GMDP and LPS slightly increase IgA in bronchoalveolar lavage; at the same time, in the asthma model, injections of GMDP or LPS before sensitization contribute to a significant decrease in IgA (2.6 times and 2.1 times, respectively) in BALF and IgE (2.2 times and 2.0 times, respectively) in blood serum. In an experimental model of asthma, the effect of GMDP and LPS was multidirectional: when they are repeatedly administered before sensitization, the bacterial components significantly reduce the severity of the allergic process, while in the case of a joint injection with an allergen, they increase the influx of macrophages, lymphocytes, and neutrophils into the lungs, which can aggravate the course of pathological process. Thus, the insufficient effect of antigens of a bacterial nature, in particular, with prolonged use of antibiotics can be compensated for by substances based on low-molecular-weight bioregulators of bacterial origin to establish the missing signals for innate immunity receptors, whose constant activation at a certain level is necessary to maintain homeostasis.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, Ministry of Science and Higher Education of the Russian Federation, 117997 Moscow, Russia
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Olga B. Gigani
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Georgii O. Gudima
- National Research Center-Institute of Immunology of the Federal Medico-Biological Agency, 115522 Moscow, Russia;
| | - Anastasiya M. Kataeva
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Natalya V. Kolesnikova
- Department of Clinical Immunology, Kuban State Medical University, Ministry of Health of the Russian Federation, 350063 Krasnodar, Russia;
| |
Collapse
|
11
|
Zhu L, Han Z, He Y, Sun H. Caspase-1-Dependent Pyroptosis Mediates Adjuvant Activity of Platycodin D as an Adjuvant for Intramuscular Vaccines. Cells 2022; 11:cells11010134. [PMID: 35011696 PMCID: PMC8750424 DOI: 10.3390/cells11010134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
Platycodin D (PD) is a potent adjuvant with dual Th1 and Th2 potentiating activity, but its mechanisms of action remain unclear. Here, the C2C12 myoblast cell line and mice were used as in vitro and in vivo models to identify potential signaling pathways involved in the adjuvant activity of PD. PD induced a transient cytotoxicity and inflammatory response in the C2C12 cells and in mouse quadricep muscles. A comparative analysis of microarray data revealed that PD induced similar gene expression profiles in the C2C12 cells and in the quadricep muscles, and triggered rapid regulation of death, immune, and inflammation-related genes, both in vivo and in vitro. It was further demonstrated that caspase-1-dependent pyroptosis was involved in the PD-induced cytotoxicity and inflammatory response in the C2C12 cells via the Ca2+–c-jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK)–NLR family pyrin domain containing 3 (NLRP3) inflammasome signaling pathway. Consistently, the in vivo analysis revealed that a local blockage of NLRP3 and caspase-1 inhibited PD-induced cytokine production and immune cell recruitment at the injection site, and impaired the adjuvant activity of PD on antigen-specific immune responses to model antigen ovalbumin (OVA) in mice. These findings identified the caspase-1-dependent adjuvanticity of PD and expanded the current knowledge on the mechanisms of action of saponin-based adjuvants.
Collapse
|
12
|
Mugisho OO, Green CR. The NLRP3 inflammasome in age-related eye disease: Evidence-based connexin hemichannel therapeutics. Exp Eye Res 2021; 215:108911. [PMID: 34958779 DOI: 10.1016/j.exer.2021.108911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/25/2021] [Accepted: 12/21/2021] [Indexed: 12/21/2022]
Abstract
The inflammasome pathway is a fundamental component of the innate immune system, playing a key role especially in chronic age-related eye diseases (AREDs). The inflammasome is of particular interest because it is a common disease pathway that once instigated, can amplify and perpetuate itself leading to chronic inflammation. With aging, it becomes more difficult to shut down inflammation after an insult but the common pathway means that a shared solution may be feasible that could be effective across multiple disease indications. This review focusses on the NLRP3 inflammasome, the most studied and characterized inflammasome in the eye. It describes the two-step signalling required for NLRP3 inflammasome complex activation, and provides evidence for its role in AREDs. In the final section, the article gives an overview of potential NLRP3 inflammasome targeting therapies, before presenting evidence for connexin hemichannel regulators as upstream blockers of inflammasome activation. These have shown therapeutic efficacy in multiple ocular disease models.
Collapse
Affiliation(s)
- Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| | - Colin R Green
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
13
|
Bayan N, Yazdanpanah N, Rezaei N. Role of Toll-Like Receptor 4 in Diabetic Retinopathy. Pharmacol Res 2021; 175:105960. [PMID: 34718133 DOI: 10.1016/j.phrs.2021.105960] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy (DR) is the most frequent microvascular complication of diabetes mellitus (DM) and a leading cause of blindness worldwide. Evidence has shown that DR is an inflammatory disease with hyperglycemia playing a causative role in the development of its main features, including inflammation, cellular apoptosis, neurodegeneration, oxidative stress, and neovascularization. Toll-like receptors (TLRs) are a well-known family of pattern recognition receptors (PRRs) responsible for the initiation of inflammatory and immune responses. TLR4 identifies both endogenous and exogenous ligands and is associated with various physiological and pathological pathways in the body. While the detailed pathophysiology of DR is still unclear, increasing data suggests a crucial role for TLR4 in the development of DR. Due to hyperglycemia, TLR4 expression increases in diabetic retina, which activates various pathways leading to DR. Considering the role of TLR4 in DR, several studies have focused on the association of TLR4 polymorphisms and risk of DR development. Moreover, evidence concerning the effect of microRNAs in the pathogenesis of DR, through their interaction with TLR4, indicates the determinant role of TLR4 in this disease. Of note, several agents have proven as effective in alleviating DR through the inhibition of the TLR4 pathway, suggesting new avenues in DR treatment. In this review, we provided a brief overview of the TLR4 structure and biological function and a more comprehensive discussion about the mechanisms of TLR4 activation in DR. Furthermore, we summarized the relationship between TLR4 polymorphisms and risk of DR and the relationship between microRNAs and TLR4 in DR. Finally, we discussed the current progress in designing TLR4 inhibitors, which could be helpful in DR clinical management.
Collapse
Affiliation(s)
- Nikoo Bayan
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Puccetti M, Gomes Dos Reis L, Pariano M, Costantini C, Renga G, Ricci M, Traini D, Giovagnoli S. Development and in vitro-in vivo performances of an inhalable indole-3-carboxaldehyde dry powder to target pulmonary inflammation and infection. Int J Pharm 2021; 607:121004. [PMID: 34391857 DOI: 10.1016/j.ijpharm.2021.121004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/17/2023]
Abstract
A tryptophan metabolite of microbial origin, indole-3-carboxaldehyde (3-IAld), has been recently identified as a Janus molecule that, acting at the host-pathogen interface and activating the aryl hydrocarbon receptor, can result as a potential candidate to treat infections as well as diseases with an inflammatory and/or immune component. In this work, an inhaled dry powder of 3-IAld was developed and evaluated for its efficacy, compared to oral and intranasal administration using an aspergillosis model of infection and inflammation. The obtained inhalable dry powder was shown to: i) be suitable to be delivered for pulmonary administration, ii) possess good toxicological safety, and iii) be superior to other administration modalities (oral and intranasal) in reducing disease scores by acting on infection and inflammation. This study supports the use of 3-IAld inhalable dry powders as a potential novel therapeutic tool to target inflammation and infection in pulmonary diseases.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy
| | - Larissa Gomes Dos Reis
- Respiratory Technology Group, The Woolcock Institute of Medical Research, Glebe, Sydney, Australia
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132 Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132 Perugia, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132 Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy
| | - Daniela Traini
- Respiratory Technology Group, The Woolcock Institute of Medical Research, Glebe, Sydney, Australia; Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy.
| |
Collapse
|
15
|
Luraghi A, Peri F, Moroni L. Electrospinning for drug delivery applications: A review. J Control Release 2021; 334:463-484. [PMID: 33781809 DOI: 10.1016/j.jconrel.2021.03.033] [Citation(s) in RCA: 244] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022]
Abstract
Drug delivery devices are promising tools in the pharmaceutical field, as they are able to maximize the therapeutic effects of the delivered drug while minimizing the undesired side effects. In the past years, electrospun nanofibers attracted rising attention due to their unique features, like biocompatibility and broad flexibility. Incorporation of active principles in nanofibrous meshes proved to be an efficient method for in situ delivery of a wide range of drugs, expanding the possibility and applicability of those devices. In this review, the principle of electrospinning and different fields of applications are treated to give an overview of the recent literature, underlining the easy tuning and endless combination of this technique, that in the future could be the new frontier of personalized medicine.
Collapse
Affiliation(s)
- Andrea Luraghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milan, Italy
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milan, Italy
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ET Maastricht, the Netherlands.
| |
Collapse
|
16
|
Jung SY, Kim D, Park DC, Lee EH, Choi YS, Ryu J, Kim SH, Yeo SG. Immunoglobulins and Transcription Factors in Otitis Media. Int J Mol Sci 2021; 22:ijms22063201. [PMID: 33801155 PMCID: PMC8004237 DOI: 10.3390/ijms22063201] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 01/13/2023] Open
Abstract
The causes of otitis media (OM) involve bacterial and viral infection, anatomo-physiological abnormalities of the Eustachian canal and nasopharynx, allergic rhinitis, group childcare centers, second-hand smoking, obesity, immaturity and defects of the immune system, formula feeding, sex, race, and age. OM is accompanied by complex and diverse interactions among bacteria, viruses, inflammatory cells, immune cells, and epithelial cells. The present study summarizes the antibodies that contribute to immune reactions in all types of otitis media, including acute otitis media, otitis media with effusion, and chronic otitis media with or without cholesteatoma, as well as the transcription factors that induce the production of these antibodies. The types and distribution of B cells; the functions of B cells, especially in otorhinolaryngology; antibody formation in patients with otitis media; and antibodies and related transcription factors are described. B cells have important functions in host defenses, including antigen recognition, antigen presentation, antibody production, and immunomodulation. The phenotypes of B cells in the ear, nose, and throat, especially in patients with otitis media, were shown to be CD5low, CD23high, CD43low, B220high, sIgMlow, sIgDhigh, Mac-1low, CD80(B7.1)low, CD86(B7.2)low, and Syndecam-1low. Of the five major classes of immunoglobulins produced by B cells, three (IgG, IgA, and IgM) are mainly involved in otitis media. Serum concentrations of IgG, IgA, and IgM are lower in patients with OM with effusion (OME) than in subjects without otitis media. Moreover, IgG, IgA, and IgM concentrations in the middle ear cavity are increased during immune responses in patients with otitis media. B cell leukemia/lymphoma-6 (Bcl-6) and paired box gene 5 (Pax-5) suppress antibody production, whereas B lymphocyte inducer of maturation program 1 (Blimp-1) and X-box binding protein 1 (XBP-1) promote antibody production during immune responses in patients with otitis media.
Collapse
Affiliation(s)
- Su Young Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, Myongji Hospital, Hanyang University College of Medicine, Goyang 10475, Korea;
| | - Dokyoung Kim
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Dong Choon Park
- Department of Gynecologic Oncology, St. Vincent’s Hospital, The Catholic University of Korea, Suwon 16247, Korea;
| | - Eun Hye Lee
- Department of Pediatrics, College of Medicine, Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.H.L.); (Y.-S.C.)
| | - Yong-Sung Choi
- Department of Pediatrics, College of Medicine, Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.H.L.); (Y.-S.C.)
| | - Jeewon Ryu
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.R.); (S.H.K.)
| | - Sang Hoon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.R.); (S.H.K.)
| | - Seung Geun Yeo
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.R.); (S.H.K.)
- Correspondence: ; Tel.: +82-2-958-8980; Fax: +82-2-958-8470
| |
Collapse
|
17
|
Han B, Hoang BX. Opinions on the current pandemic of COVID-19: Use functional food to boost our immune functions. J Infect Public Health 2020; 13:1811-1817. [PMID: 32948484 PMCID: PMC7831995 DOI: 10.1016/j.jiph.2020.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022] Open
Abstract
The pandemic of novel coronavirus caused COVID-19 had resulted in a high number of hospitalizations and deaths and caused a devastating toll on human and society health. The symptoms of the infected patients vary significantly, from life-threatening to mild or even asymptomatic. This clinical observation led to hypothesize on the critical role of host innate immunity in the disease development and progression. As the first defense barrier against microorganisms, the innate immune reaction determines not only the viral infection rate but also immune-mediated response. Therefore, promote healthy behaviors to enhance innate immunity with functional food and nutritional agents may be a rational strategy for minimizing damages caused by viruses to global health.
Collapse
Affiliation(s)
- Bo Han
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States
| | - Ba X Hoang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
18
|
Puccetti M, Xiroudaki S, Ricci M, Giovagnoli S. Postbiotic-Enabled Targeting of the Host-Microbiota-Pathogen Interface: Hints of Antibiotic Decline? Pharmaceutics 2020; 12:E624. [PMID: 32635461 PMCID: PMC7408102 DOI: 10.3390/pharmaceutics12070624] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Mismanagement of bacterial infection therapies has undermined the reliability and efficacy of antibiotic treatments, producing a profound crisis of the antibiotic drug market. It is by now clear that tackling deadly infections demands novel strategies not only based on the mere toxicity of anti-infective compounds. Host-directed therapies have been the first example as novel treatments with alternate success. Nevertheless, recent advances in the human microbiome research have provided evidence that compounds produced by the microbial metabolism, namely postbiotics, can have significant impact on human health. Such compounds target the host-microbe-pathogen interface rescuing biotic and immune unbalances as well as inflammation, thus providing novel therapeutic opportunities. This work discusses critically, through literature review and personal contributions, these novel nonantibiotic treatment strategies for infectious disease management and resistance prevention, which could represent a paradigm change rocking the foundation of current antibiotic therapy tenets.
Collapse
Affiliation(s)
| | | | | | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, via del Liceo 1, University of Perugia, 06123 Perugia, Italy; (M.P.); (S.X.); (M.R.)
| |
Collapse
|
19
|
Lu BL, Williams GM, Brimble MA. TLR2 agonists and their structure–activity relationships. Org Biomol Chem 2020; 18:5073-5094. [DOI: 10.1039/d0ob00942c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We review the structure–activity relationships and synthetic studies of TLR2 agonists – important chemical targets in immunotherapy.
Collapse
Affiliation(s)
- Benjamin L. Lu
- The School of Biological Sciences
- University of Auckland
- Auckland 1010
- New Zealand
- The School of Chemical Sciences
| | - Geoffrey M. Williams
- The School of Biological Sciences
- University of Auckland
- Auckland 1010
- New Zealand
- The School of Chemical Sciences
| | - Margaret A. Brimble
- The School of Biological Sciences
- University of Auckland
- Auckland 1010
- New Zealand
- The School of Chemical Sciences
| |
Collapse
|
20
|
Lei JR, Tu XK, Wang Y, Tu DW, Shi SS. Resveratrol downregulates the TLR4 signaling pathway to reduce brain damage in a rat model of focal cerebral ischemia. Exp Ther Med 2019; 17:3215-3221. [PMID: 30936996 DOI: 10.3892/etm.2019.7324] [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] [Received: 06/23/2018] [Accepted: 11/23/2018] [Indexed: 12/21/2022] Open
Abstract
Previous studies have demonstrated that inflammation and disruption of the blood-brain barrier (BBB) are important pathological processes during focal cerebral ischemia. Therefore, the present study evaluated the neuroprotective effects of resveratrol against brain damage, inflammation and BBB disruption in rats with focal cerebral ischemia and assessed the potential underlying molecular mechanisms. Sprague-Dawley rats underwent cerebral ischemia/reperfusion (IR) and then received intraperitoneal resveratrol (10 and 100 mg/kg) 2 h following the onset of ischemia. Following 24 h of ischemia, neurological deficit scores, cerebral infarctions, morphological characteristics, cerebral water content, myeloperoxidase (MPO) activity and Evans blue extravasation were assessed. Additionally, the protein expression levels of Toll-like receptor 4 (TLR4) and nuclear factor (NF)-κB p65 were detected using western blot analyses, the mRNA expression levels of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were examined by reverse-transcription polymerase chain reaction, and tumor necrosis factor (TNF)-α and interleukin (IL)-1β blood levels were determined by ELISA. Resveratrol significantly reduced neurological deficit scores, cerebral infarct sizes, neuronal injury, MPO activity and EB content. Cerebral ischemia increased the expression levels of TLR4, NF-κB p65, COX-2, MMP-9, TNF-α and IL-1β, but all of these factors were reduced by resveratrol. In conclusion, the present data suggest that resveratrol reduces inflammation, BBB disruption and brain damage in rats following focal cerebral ischemia. Additionally, the neuroprotective effects of resveratrol against cerebral ischemia may be associated with downregulation of the TLR4 pathway.
Collapse
Affiliation(s)
- Jun-Rong Lei
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yang Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - De-Wen Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
21
|
Gupta PK, Kulkarni S. Polysaccharide rich extract (PRE) from Tinospora cordifolia inhibits the intracellular survival of drug resistant strains of Mycobacterium tuberculosis in macrophages by nitric oxide induction. Tuberculosis (Edinb) 2018; 113:81-90. [PMID: 30514517 DOI: 10.1016/j.tube.2018.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/23/2018] [Accepted: 09/23/2018] [Indexed: 11/15/2022]
Abstract
Plethora of clinical and scientific information obtained in recent past has strengthened the idea that targeting critical constituents of host immune system may have beneficial outcomes for the treatment of tuberculosis. Macrophages being the primary host for Mycobacterium tuberculosis, offer an attractive target for modulation. Owing to their negligible toxicity, plant derived polysaccharides with the ability to activate macrophages; are suitable candidates for immunomodulation. In the present study, effects of polysaccharide rich extract (PRE) isolated from Tinospora cordifolia, on the survival of intracellular MTB strains and activation of macrophages were investigated. PRE treatment up regulated the expression of pro-inflammatory cytokines such as IL-β, TNF-α, IL-6, IL-12, and IFN-γ in RAW 264.7 cell line. Up regulation in the expression of NOS2 was observed along with concomitant enhanced nitric oxide production post PRE treatment. Surface expression of MHC-II and CD-86 was up regulated after PRE treatment. Above results suggested the classical activation of macrophages by PRE treatment. Furthermore, PRE treatment led to the activation of all the three classes of MAPK i.e p38, ERK and JNK MAPKs. Further, PRE up regulated the expression of cytokines, NOS-2, MHC-II and CD-86 in MTB infected macrophages. PRE treatment inhibited the intracellular survival of drug resistant MTB in macrophages which was partially attributed to PRE mediated NO induction. Thus our data demonstrate classical activation of macrophages by PRE treatment and killing of intracellular MTB by NO induction.
Collapse
Affiliation(s)
- Pramod Kumar Gupta
- Radiation Medicine Centre, Bhabha Atomic Research Centre, c/o TMH Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India.
| | - Savita Kulkarni
- Radiation Medicine Centre, Bhabha Atomic Research Centre, c/o TMH Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India.
| |
Collapse
|
22
|
Kim H, Sehgal D, Kucaba TA, Ferguson DM, Griffith TS, Panyam J. Acidic pH-responsive polymer nanoparticles as a TLR7/8 agonist delivery platform for cancer immunotherapy. NANOSCALE 2018; 10:20851-20862. [PMID: 30403212 DOI: 10.1039/c8nr07201a] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Synthetic imidazoquinoline-based toll-like receptor (TLR) 7/8 bi-specific agonists are promising vaccine adjuvants that can induce maturation of dendritic cells (DCs) and activate them to secrete pro-inflammatory cytokines. However, in vivo efficacy of these small molecule agonists is often hampered by their fast clearance from the injection site, limiting their use to topical treatments. In this study, we investigated the use of acidic pH-responsive poly(lactide-co-glycolide) (PLGA) nanoparticles for endo-lysosome specific release of 522, a novel TLR7/8 agonist. Bicarbonate salt was incorporated into the new formulation to generate carbon dioxide (CO2) gas at acidic pH, which can disrupt the polymer shell to rapidly release the payload. Compared to conventional PLGA nanoparticles, the pH responsive formulation resulted in 33-fold higher loading of 522. The new formulation demonstrated acid-responsive CO2 gas generation and drug release. The acid-responsive formulation increased the in vitro expression of co-stimulatory molecules on DCs and improved antigen-presentation via MHC I, both of which are essential for CD8 T cell priming. In vivo studies showed that the pH-responsive formulation elicited stronger antigen-specific CD8 T cell and natural killer (NK) cell responses than conventional PLGA nanoparticles, resulting in enhanced anticancer efficacy in a murine melanoma tumor model. Our results suggest that acidic-pH responsive, gas-generating nanoparticles are an efficient TLR7/8 agonist delivery platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Hyunjoon Kim
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Li P, Rios Coronado PE, Longstaff XRR, Tarashansky AJ, Wang B. Nanomedicine Approaches Against Parasitic Worm Infections. Adv Healthc Mater 2018; 7:e1701494. [PMID: 29602254 DOI: 10.1002/adhm.201701494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/01/2018] [Indexed: 01/10/2023]
Abstract
Nanomedicine approaches have the potential to transform the battle against parasitic worm (helminth) infections, a major global health scourge from which billions are currently suffering. It is anticipated that the intersection of two currently disparate fields, nanomedicine and helminth biology, will constitute a new frontier in science and technology. This progress report surveys current innovations in these research fields and discusses research opportunities. In particular, the focus is on: (1) major challenges that helminth infections impose on mankind; (2) key aspects of helminth biology that inform future research directions; (3) efforts to construct nanodelivery platforms to target drugs and genes to helminths hidden in their hosts; (4) attempts in applying nanotechnology to enable vaccination against helminth infections; (5) outlooks in utilizing nanoparticles to enhance immunomodulatory activities of worm-derived factors to cure allergy and autoimmune diseases. In each section, achievements are summarized, limitations are explored, and future directions are assessed.
Collapse
Affiliation(s)
- Pengyang Li
- Department of Bioengineering; Stanford University; Stanford CA 94305 USA
| | | | | | | | - Bo Wang
- Department of Bioengineering; Stanford University; Stanford CA 94305 USA
| |
Collapse
|
24
|
Fu J, van Wietmarschen HA, van der Kooij A, Cuppen BV, Schroën Y, Marijnissen AK, Meulman JJ, Lafeber FP, van der Greef J. Systems approach for classifying the response to biological therapies in patients with rheumatoid arthritis in clinical practice. Eur J Integr Med 2018. [DOI: 10.1016/j.eujim.2018.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
25
|
Elburki MS, Rossa C, Guimarães-Stabili MR, Lee HM, Curylofo-Zotti FA, Johnson F, Golub LM. A Chemically Modified Curcumin (CMC 2.24) Inhibits Nuclear Factor κB Activation and Inflammatory Bone Loss in Murine Models of LPS-Induced Experimental Periodontitis and Diabetes-Associated Natural Periodontitis. Inflammation 2018; 40:1436-1449. [PMID: 28534138 DOI: 10.1007/s10753-017-0587-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The purpose of this study was to assess the effect of a novel chemically modified curcumin (CMC 2.24) on NF-κB and MAPK signaling and inflammatory cytokine production in two experimental models of periodontal disease in rats. Experimental model I: Periodontitis was induced by repeated injections of LPS into the gingiva (3×/week, 3 weeks); control rats received vehicle injections. CMC 2.24, or the vehicle, was administered by daily oral gavage for 4 weeks. Experimental model II: Diabetes was induced in adult male rats by streptozotocin injection; periodontal breakdown then results as a complication of uncontrolled hyperglycemia. Non-diabetic rats served as controls. CMC 2.24, or the vehicle, was administered by oral gavage daily for 3 weeks to the diabetics. Hemimaxillae and gingival tissues were harvested, and bone loss was assessed radiographically. Gingival tissues were pooled according to the experimental conditions and processed for the analysis of matrix metalloproteinases (MMPs) and bone-resorptive cytokines. Activation of p38 MAPK and NF-κB signaling pathways was assessed by western blot. Both LPS and diabetes induced an inflammatory process in the gingival tissues associated with excessive alveolar bone resorption and increased activation of p65 (NF-κB) and p38 MAPK. In both models, the administration of CMC 2.24 produced a marked reduction of inflammatory cytokines and MMPs in the gingival tissues, decreased bone loss, and decreased activation of p65 (NF-κB) and p38 MAPK. Inhibition of these cell signaling pathways by this novel tri-ketonic curcuminoid (natural curcumin is di-ketonic) may play a role in its therapeutic efficacy in locally and systemically associated periodontitis.
Collapse
Affiliation(s)
- Muna S Elburki
- Department of Periodontics, Faculty of Dentistry, University of Benghazi, Jamal Abdel Nasser Street, Benghazi, Libya.
| | - Carlos Rossa
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara-UNESP, Araraquara, Brazil
| | | | - Hsi-Ming Lee
- Department of Oral Biology and Pathology, School of Dental Medicine, SUNY at Stony Brook, Stony Brook, NY, USA
| | - Fabiana A Curylofo-Zotti
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara-UNESP, Araraquara, Brazil
| | - Francis Johnson
- Department of Chemistry and Pharmacological Sciences, SUNY at Stony Brook, Stony Brook, NY, USA
| | - Lorne M Golub
- Department of Oral Biology and Pathology, School of Dental Medicine, SUNY at Stony Brook, Stony Brook, NY, USA
| |
Collapse
|
26
|
High-Throughput Screening for Identification of Novel Innate Immune Activators. Methods Mol Biol 2017. [PMID: 28808971 DOI: 10.1007/978-1-4939-7237-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Modern drug discovery has embraced in vitro platforms that enable investigation of large numbers of compounds within tractable timeframes and for feasible costs. These endeavors have been greatly aided in recent years by advances in molecular and cell-based methods such as gene delivery and editing technology, advanced imaging, robotics, and quantitative analysis. As such, the examination of phenotypic impacts of novel molecules may only be limited by the size of the compound collection. Innate immune signaling processes in mammalian cells are especially amenable to high-throughput screening platforms since the cellular responses elicited by their activation often result in high level transcription that can be harnessed in the form of bioluminescent or fluorescent signal. In addition, targeted activation of innate immune pathways represents a valuable therapeutic strategy applicable to multiple chronic and acute human diseases. Herein, we describe the optimization and utilization of a high-throughput screening method using human reporter cells reactive to stimulation of the type I interferon response. Importantly, the principles and methods described can be applied to adherent reporter cells of diverse derivation and innate signaling pathway readouts.
Collapse
|
27
|
Farrokhi S, Abbasirad N, Movahed A, Khazaei HA, Pishjoo M, Rezaei N. TLR9-based immunotherapy for the treatment of allergic diseases. Immunotherapy 2017; 9:339-346. [PMID: 28303762 DOI: 10.2217/imt-2016-0104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Toll-like receptors (TLRs), a family of pattern recognition receptors expressed on many cell types of innate immunity, recognize the pathogen-associated molecular patterns of microbes. The hygiene hypothesis suggests that a reduced microbial exposure in early childhood increases the susceptibility to allergic diseases due to deviation in development of the immune system. TLRs are key roles in the right and healthy direction of adaptive immunity with the induction of T-helper 2 toward Th1 immune responses and regulatory T cells. TLR ligand CpG-ODN-based immunomodulation is independent of allergen and it mainly affects innate immune system. While, CpG-oligodeoxynucleotide-based vaccination is allergen specific and induces adaptive immune system. The use of agonists of TLR9 in two distinct strategies of immunotherapy, immunomodulation and vaccination, could be presented as the curative method for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Shokrollah Farrokhi
- Department of Immunology, Asthma & Allergy, The Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Narjes Abbasirad
- Department of Immunology, Asthma & Allergy, The Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Movahed
- Department of Biochemistry, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hossein Ali Khazaei
- Clinical Immunology Research Center, Department of Immunology & Hematology, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Pishjoo
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Eichinger KM, Resetar E, Orend J, Anderson K, Empey KM. Age predicts cytokine kinetics and innate immune cell activation following intranasal delivery of IFNγ and GM-CSF in a mouse model of RSV infection. Cytokine 2017; 97:25-37. [PMID: 28558308 DOI: 10.1016/j.cyto.2017.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/20/2017] [Accepted: 05/23/2017] [Indexed: 12/22/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in young children and is further associated with increased healthcare utilization and cost of care in the first years of life. Severe RSV disease during infancy has also been linked to the later development of allergic asthma, yet there remains no licensed RSV vaccine or effective treatment. Pre-clinical and clinical studies have shown that disease severity and development of allergic asthma are associated with differences in cytokine production. As a result, stimulation of the innate host immune response with immune potentiators is gaining attention for their prospective application in populations with limited immune responses to antigenic stimuli or against pathogens for which vaccines do not exist. Specifically, macrophage-activating cytokines such as interferon gamma (IFNγ) and granulocyte colony-stimulating factor (GM-CSF) are commercially available immune potentiators used to prevent infections in patients with chronic granulomatous disease and febrile neutropenia, respectively. Moreover, an increasing number of reports describe the protective function of IFNγ and GM-CSF as vaccine adjuvants. Although a positive correlation between cytokine production and age has previously been reported, little is known about age-dependent cytokine metabolism or immune activating responses in infant compared to adult lungs. Here we use a non-compartmental pharmacokinetic model in naïve and RSV-infected infant and adult BALB/c mice to determine the effect of age on IFNγ and GM-CSF elimination and innate cell activation following intranasal delivery.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Erin Resetar
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacob Orend
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Kacey Anderson
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Cho WK, Jang JP, Choi EJ, Ahn M, Kim SH, Cho KS, Park SH, Baek IC, Jung MH, Kim TG, Suh BK. Association of Polymorphisms in Toll-Like Receptors 4 and 9 with Autoimmune Thyroid Disease in Korean Pediatric Patients. Int J Endocrinol 2017; 2017:2304218. [PMID: 28912808 PMCID: PMC5585642 DOI: 10.1155/2017/2304218] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/13/2017] [Accepted: 07/25/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Toll-like receptors (TLRs) have been suggested to be associated with the development of AITD. METHODS Fifteen single-nucleotide polymorphisms in 7 TLR genes were analyzed in 104 Korean children (girls = 86, boys = 18) with AITD (Hashimoto disease (HD) = 44, Graves' disease (GD) = 60, thyroid-associated ophthalmopathy (TAO) = 29, and non-TAO = 31) with 183 controls. RESULTS GD showed higher frequencies of the TLR4 rs1927911 C allele than control. TAO showed a lower frequency of the TLR4 rs1927911 CT genotype and non-TAO showed a higher frequency of the TLR4 rs1927911 CC genotype than control. The frequency of the TLR9 rs187084 CC genotype in TAO was higher than that in non-TAO. GD females showed a higher frequency of the TLR4 rs10759932 T allele, rs1927911 CC genotype, and the rs1927911 C allele than controls. GD males showed a higher frequency of the TLR4 rs10759932 CC genotype and rs1927911 TT genotype and lower frequency of the rs1927911 CT genotype than control. The frequency of the TLR4 rs10759932 CC genotype, C allele and rs1927911 TT genotype, and T allele in a GD female were lower than in a GD male. CONCLUSIONS Our results suggest that TLR4 and 9 polymorphisms might contribute to the pathogenesis of GD and TAO.
Collapse
Affiliation(s)
- Won Kyoung Cho
- Department of Pediatrics, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Pil Jang
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun-Jeong Choi
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Moonbae Ahn
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Shin Hee Kim
- Department of Pediatrics, College of Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoung Soon Cho
- Department of Pediatrics, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - So Hyun Park
- Department of Pediatrics, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - In Cheol Baek
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Ho Jung
- Department of Pediatrics, College of Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- *Tai-Gyu Kim: and
| | - Byung-Kyu Suh
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- *Byung-Kyu Suh:
| |
Collapse
|
30
|
Nawaz M, Fatima F, Nazarenko I, Ekström K, Murtaza I, Anees M, Sultan A, Neder L, Camussi G, Valadi H, Squire JA, Kislinger T. Extracellular vesicles in ovarian cancer: applications to tumor biology, immunotherapy and biomarker discovery. Expert Rev Proteomics 2016; 13:395-409. [PMID: 26973172 DOI: 10.1586/14789450.2016.1165613] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years there has been tremendous interest in both the basic biology and applications of extracellular vesicles (EVs) in translational cancer research. This includes a better understanding of their biogenesis and mechanisms of selective cargo packaging, their precise roles in horizontal communication, and their application as non-invasive biomarkers. The rapid advances in next-generation omics technologies are the driving forces for these discoveries. In this review, the authors focus on recent results of EV research in ovarian cancer. A deeper understanding of ovarian cancer-derived EVs, the types of cargo molecules and their biological roles in cancer growth, metastases and drug resistance, could have significant impact on the discovery of novel biomarkers and innovative therapeutics. Insights into the role of EVs in immune regulation could lead to novel approaches built on EV-based immunotherapy.
Collapse
Affiliation(s)
- Muhammad Nawaz
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Farah Fatima
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Irina Nazarenko
- c Institute for Environmental Health Sciences and Hospital Infection Control , University Medical Centre Freiburg , Freiburg im Breisgau , Germany
| | - Karin Ekström
- d Department of Biomaterials , Sahlgrenska Academy at the University of Gothenburg , Gothenburg , Sweden.,e BIOMATCELL VINN Excellence Centre of Biomaterials and Cell Therapy , Gothenburg , Sweden
| | - Iram Murtaza
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Mariam Anees
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Aneesa Sultan
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Luciano Neder
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Giovanni Camussi
- g Department of Medical Sciences and Molecular Biotechnology Centre , University of Torino , Torino , Italy
| | - Hadi Valadi
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Jeremy A Squire
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Thomas Kislinger
- h Princess Margaret Cancer Centre and Department of Medical Biophysics , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
31
|
Peng S, Li C, Wang X, Liu X, Han C, Jin T, Liu S, Zhang X, Zhang H, He X, Xie X, Yu X, Wang C, Shan L, Fan C, Shan Z, Teng W. Increased Toll-Like Receptors Activity and TLR Ligands in Patients with Autoimmune Thyroid Diseases. Front Immunol 2016; 7:578. [PMID: 28018345 PMCID: PMC5145898 DOI: 10.3389/fimmu.2016.00578] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/24/2016] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Autoimmune thyroid disease (AITD) is an organ-specific disorder due to the interplay between environmental and genetic factors. Toll-like receptors (TLRs) are pattern recognition receptors expressed abundantly on monocytes. There is a paucity of data on TLR expression in AITD. The aim of this study was to examine TLR expression, activation, ligands, and downstream signaling adaptors in peripheral blood mononuclear cells (PBMCs) extracted from untreated AITD patients and healthy controls. METHOD We isolated PBMC of 30 healthy controls, 36 patients with untreated Hashimoto's thyroiditis, and 30 patients with newly onset Graves' disease. TLR mRNA, protein expression, TLR ligands, and TLR adaptor molecules were measured using real-time PCR, Western blot, flow cytometry, and enzyme-linked immunosorbent assay (ELISA). PBMC was simulated with TLR agonists. The effects of TLR agonists on the viability of human PBMC were evaluated using the MTT assay. The supernatants of cell cultures were measured for the pro-inflammatory cytokines, interleukin (IL)-6, tumor necrosis factor alpha (TNF-α), and IL-10 by ELISA. RESULTS TLR2, TLR3, TLR9, and TLR10 mRNA were significantly increased in AITD patients compared with controls. TLR2, TLR3, TLR9, high mobility group box 1 (HMGB1), and RAGE expression on monocytes was higher in patients than control at baseline and TLR agonists' stimulation. The release of TNF-α and IL-6 was significantly increased in PBMCs from AITD patients with TLR agonists, while IL-10 was significantly decreased. Downstream targets of TLR, myeloid differentiation factor 88 (MyD88), and myeloid toll/IL-1 receptor-domain containing adaptor-inducing interferon-β were significantly elevated in AITD patients. Levels of TLR2 ligands, HMGB1, and heat shock protein 60 were significantly elevated in AITD patients compared with those in controls and positively correlated with TgAb and TPOAb, while sRAGE concentration was significantly decreased in AITD patients. CONCLUSION This work is the first to show that TLR2, TLR3, and TLR9 expression and activation are elevated in the PBMCs of patients with AITD and TLRs may participate in the pathogenesis of AITD.
Collapse
Affiliation(s)
- Shiqiao Peng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Chenyan Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology and Metabolism, The First Hospital of China Medical University, Shenyang, China
| | - Xinyi Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xin Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Intensive Care Unit, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cheng Han
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Ting Jin
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology, Sir Run Run Shaw Hospital, Affiliated to School of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Emergency, People’s Liberation Army No.202 Hospital, Shenyang, China
| | - Xiaowen Zhang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Hanyi Zhang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Xue He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Xiaochen Xie
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Xiaohui Yu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology and Metabolism, The First Hospital of China Medical University, Shenyang, China
| | - Chuyuan Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology and Metabolism, The First Hospital of China Medical University, Shenyang, China
| | - Ling Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Chenling Fan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology and Metabolism, The First Hospital of China Medical University, Shenyang, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China,Department of Endocrinology and Metabolism, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
32
|
Zakaria S, Mahmoud AA, Hasan RA, Mahmoud MF, El Fayoumi HM. Cinnamaldehyde Mitigates Carbon Tetrachloride-induced Acute Liver Injury in Rats Through Inhibition of Toll-like Receptor 4 Signaling Pathway. INT J PHARMACOL 2016. [DOI: 10.3923/ijp.2016.851.862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
33
|
Vu A, Calzadilla A, Gidfar S, Calderon-Candelario R, Mirsaeidi M. Toll-like receptors in mycobacterial infection. Eur J Pharmacol 2016; 808:1-7. [PMID: 27756604 DOI: 10.1016/j.ejphar.2016.10.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 10/04/2016] [Accepted: 10/14/2016] [Indexed: 10/20/2022]
Abstract
Toll-like receptors are transmembrane glycoproteins predominantly expressed in tissues with immune function. They are considered one of the most important pattern recognition receptor families discovered at the end of 20th century and a key aspect of the innate immune system response to infectious disease. Here we present a review of the current knowledge of individual Toll-like receptors, 1 through 13, with a focus on their role in the immune system response to mycobacterial infection. We present literature to date about the Toll-like receptors structure, localization and expression, signaling pathways, and function. The Toll-like receptor family may have proven an important role in the immune system response to mycobacterial infections, including M. tuberculosis and non-tuberculous (NTM) organisms.
Collapse
Affiliation(s)
- Ann Vu
- Department of Medicine, University of Miami, 1600 NW, Miami, FL 33136, USA.
| | - Andrew Calzadilla
- Department of Medicine, University of Miami, 1600 NW, Miami, FL 33136, USA.
| | - Sanaz Gidfar
- Department of Medicine, University of Miami, 1600 NW, Miami, FL 33136, USA.
| | - Rafael Calderon-Candelario
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW, Miami, FL 33136, USA; Miami VA Medical Center, 1201 N.W. 16th St., Miami, FL 33125, USA.
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW, Miami, FL 33136, USA; Miami VA Medical Center, 1201 N.W. 16th St., Miami, FL 33125, USA.
| |
Collapse
|
34
|
Torices S, Julia A, Muñoz P, Varela I, Balsa A, Marsal S, Fernández-Nebro A, Blanco F, López-Hoyos M, Martinez-Taboada V, Fernández-Luna JL. A functional variant of TLR10 modifies the activity of NFkB and may help predict a worse prognosis in patients with rheumatoid arthritis. Arthritis Res Ther 2016; 18:221. [PMID: 27716427 PMCID: PMC5050569 DOI: 10.1186/s13075-016-1113-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/06/2016] [Indexed: 02/06/2023] Open
Abstract
Background Toll-like receptor (TLR) family members are key players in inflammation. TLR10 has been poorly studied in chronic inflammatory disorders, and its clinical relevance in rheumatoid arthritis (RA) is as yet unknown. We aimed at identifying TLR10 variants within all coding regions of the gene in patients with RA as well as studying their functional and clinical significance. Methods TLR10 gene variants were studied by performing sequencing of 66 patients with RA and 30 control subjects. A selected variant, I473T, was then analyzed in 1654 patients and 1702 healthy control subjects. The capacity of this TLR10 variant to modify the transcriptional activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) was determined by using a luciferase reporter assay and analyzing the expression of NFkB target genes by quantitative polymerase chain reaction. Differences between groups were analyzed by using the Mann-Whitney U test and the unpaired two-tailed Student’s t test. Results We detected ten missense variants in the TLR10 gene and focused on the I473T substitution based on allele frequencies and the predicted functional impact. I473T variant is not associated with susceptibility to RA, but it significantly correlates with erosive disease in patients seropositive for antibodies to citrullinated protein antigens (p = 0.017 in the total cohort and p = 0.0049 in female patients) and with a lower response to infliximab treatment as measured by the change in Disease Activity Score in 28 joints (p = 0.012) and by the European League Against Rheumatism criteria (p = 0.049). Functional studies showed that TLR10 reduced activation of the NFkB inflammatory pathway in hematopoietic cells, whereas the I473T variant lacked this inhibitory capacity. Consistently, after exposure to infliximab, cells expressing the I437T variant showed higher NFkB activity than cells carrying wild-type TLR10. Conclusions A TLR10 allelic variant, I473T, has impaired NFkB inhibitory activity and is highly associated with disease severity and low response to infliximab in patients with RA.
Collapse
Affiliation(s)
- Silvia Torices
- Servicio de Reumatología, Hospital Universitario Marques de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Avenida Valdecilla s/n, 39008, Santander, Spain.,Unidad de Genética, Hospital Universitario Marques de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Avenida Valdecilla s/n, 39008, Santander, Spain
| | - Antonio Julia
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Pedro Muñoz
- Gerencia Atención Primaria, Servicio Cántabro de Salud, 39011, Santander, Spain
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, 39011, Santander, Spain
| | - Alejandro Balsa
- Servicio de Reumatología, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Antonio Fernández-Nebro
- Unidad de Reumatología, Instituto de Investigación Biomédica de Málaga, Hospital Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Francisco Blanco
- Departamento de Reumatología, Hospital Universitario A Coruña, 15006, A Coruña, Spain
| | - Marcos López-Hoyos
- Sección de Inmunología, Hospital Universitario Marques de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Avenida Valdecilla s/n, 39008, Santander, Spain
| | - Víctor Martinez-Taboada
- Servicio de Reumatología, Hospital Universitario Marques de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Avenida Valdecilla s/n, 39008, Santander, Spain.,Facultad de Medicina, Universidad de Cantabria, 39011, Santander, Spain
| | - Jose L Fernández-Luna
- Unidad de Genética, Hospital Universitario Marques de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Avenida Valdecilla s/n, 39008, Santander, Spain.
| |
Collapse
|
35
|
Abstract
Animals that cannot sense endotoxin may die if they are infected by Gram-negative bacteria. Animals that sense endotoxin and respond too vigorously may also die, victims of their own inflammatory reactions. The outcome of Gram-negative bacterial infection is thus determined not only by an individual's ability to sense endotoxin and respond to its presence, but also by numerous phenomena that inactivate endotoxin and/or prevent harmful reactions to it. Endotoxin sensing requires the MD-2/TLR4 recognition complex and occurs principally in local tissues and the liver. This review highlights the known detoxification mechanisms, which include: (i) proteins that facilitate LPS sequestration by plasma lipoproteins, prevent interactions between the bioactive lipid A moiety and MD-2/TLR4, or promote cellular uptake via non-signaling pathway(s); (ii) enzymes that deacylate or dephosphorylate lipid A; (iii) mechanisms that remove LPS and Gram-negative bacteria from the bloodstream; and (iv) neuroendocrine adaptations that modulate LPS-induced mediator production or neutralize pro-inflammatory molecules in the circulation. In general, the mechanisms for sensing and detoxifying endotoxin seem to be compartmentalized (local versus systemic), dynamic, and variable between individuals. They may have evolved to confine infection and inflammation to extravascular sites of infection while preventing harmful systemic reactions. Integration of endotoxin sensing and detoxification is essential for successful host defense.
Collapse
Affiliation(s)
- Robert S. Munford
- Molecular Host Defense Laboratory, Departments of Internal Medicine and Microbiology, University of Texas Southwestern Medical School, Dallas, Texas, USA,
| |
Collapse
|
36
|
Yu D, Wu Y, Xu L, Fan Y, Peng L, Xu M, Yao YG. Identification and characterization of toll-like receptors (TLRs) in the Chinese tree shrew (Tupaia belangeri chinensis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:127-138. [PMID: 26923770 DOI: 10.1016/j.dci.2016.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/23/2016] [Indexed: 06/05/2023]
Abstract
In mammals, the toll-like receptors (TLRs) play a major role in initiating innate immune responses against pathogens. Comparison of the TLRs in different mammals may help in understanding the TLR-mediated responses and developing of animal models and efficient therapeutic measures for infectious diseases. The Chinese tree shrew (Tupaia belangeri chinensis), a small mammal with a close relationship to primates, is a viable experimental animal for studying viral and bacterial infections. In this study, we characterized the TLRs genes (tTLRs) in the Chinese tree shrew and identified 13 putative TLRs, which are orthologs of mammalian TLR1-TLR9 and TLR11-TLR13, and TLR10 was a pseudogene in tree shrew. Positive selection analyses using the Maximum likelihood (ML) method showed that tTLR8 and tTLR9 were under positive selection, which might be associated with the adaptation to the pathogen challenge. The mRNA expression levels of tTLRs presented an overall low and tissue-specific pattern, and were significantly upregulated upon Hepatitis C virus (HCV) infection. tTLR4 and tTLR9 underwent alternative splicing, which leads to different transcripts. Phylogenetic analysis and TLR structure prediction indicated that tTLRs were evolutionarily conserved, which might reflect an ancient mechanism and structure in the innate immune response system. Taken together, TLRs had both conserved and unique features in the Chinese tree shrew.
Collapse
Affiliation(s)
- Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Li Peng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
37
|
D'Alonzo D, Cipolletti M, Tarantino G, Ziaco M, Pieretti G, Iadonisi A, Palumbo G, Alfano A, Giuliano M, De Rosa M, Schiraldi C, Cammarota M, Parrilli M, Bedini E, Corsaro MM. A Semisynthetic Approach to New Immunoadjuvant Candidates: Site-Selective Chemical Manipulation ofEscherichia coliMonophosphoryl Lipid A. Chemistry 2016; 22:11053-63. [DOI: 10.1002/chem.201601284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Indexed: 01/25/2023]
Affiliation(s)
- Daniele D'Alonzo
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Manuela Cipolletti
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
- Department of Biology; University “Roma Tre”; Viale G. Marconi 446 00146 Rome Italy
| | - Giulia Tarantino
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
- Cardiff Catalysis Institute; School of Chemistry; Cardiff University; Main Building, Park Place CF10 3AT Cardiff The United Kingdom
| | - Marcello Ziaco
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Giuseppina Pieretti
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Alfonso Iadonisi
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Giovanni Palumbo
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Alberto Alfano
- Department of Experimental Medicine; Second University of Naples; via de Crecchio 7 80138 Naples Italy
| | - Mariateresa Giuliano
- Department of Experimental Medicine; Second University of Naples; via de Crecchio 7 80138 Naples Italy
| | - Mario De Rosa
- Department of Experimental Medicine; Second University of Naples; via de Crecchio 7 80138 Naples Italy
| | - Chiara Schiraldi
- Department of Experimental Medicine; Second University of Naples; via de Crecchio 7 80138 Naples Italy
| | - Marcella Cammarota
- Department of Experimental Medicine; Second University of Naples; via de Crecchio 7 80138 Naples Italy
| | - Michelangelo Parrilli
- Department of Biology; University of Naples Federico II; Complesso Universitario Monte S. Angelo via Cintia 4 80126 Naples Italy
| | - Emiliano Bedini
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| | - Maria M. Corsaro
- Department of Chemical Sciences; University of Naples Federico II; Complesso Universitario Monte S. Angelo, via Cintia 4 80126 Naples Italy
| |
Collapse
|
38
|
Gupta PK, Chakraborty P, Kumar S, Singh PK, Rajan MGR, Sainis KB, Kulkarni S. G1-4A, a Polysaccharide from Tinospora cordifolia Inhibits the Survival of Mycobacterium tuberculosis by Modulating Host Immune Responses in TLR4 Dependent Manner. PLoS One 2016; 11:e0154725. [PMID: 27148868 PMCID: PMC4858241 DOI: 10.1371/journal.pone.0154725] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/18/2016] [Indexed: 11/19/2022] Open
Abstract
Rapid emergence of drug resistance in Mycobacterium tuberculosis (MTB) is a major health concern and demands the development of novel adjunct immunotherapeutic agents capable of modulating the host immune responses in order to control the pathogen. In the present study, we sought to investigate the immunomodulatory effects of G1-4A, a polysaccharide derived from the Indian medicinal plant Tinospora cordifolia, in in-vitro and aerosol mouse models of MTB infection. G1-4A treatment of MTB infected RAW264.7 macrophages significantly induced the surface expression of MHC-II and CD-86 molecules, secretion of proinflammatory cytokines (TNF-α, IL-β, IL-6, IL-12, IFN-γ) and nitric oxide leading to reduced intracellular survival of both drug sensitive (H37Rv) as well as multi drug resistant strains (Beijing and LAM) of MTB, which was partially attributed to G1-4A induced NO production in TLR4-MyD88 dependent manner. Similarly, bacillary burden was significantly reduced in the lungs of MTB infected BALB/c mice treated with G1-4A, with simultaneous up-regulation of the expression of TNF-α, INF-γ and NOS2 in the mouse lung along with increased levels of Th1 cytokines like IFN-γ, IL-12 and decreased levels of Th2 cytokine like IL-4 in the serum. Furthermore, combination of G1-4A with Isoniazid (INH) exhibited better protection against MTB compared to that due to INH or G1-4A alone, suggesting its potential as adjunct therapy. Our results demonstrate that modulation of host immune responses by G1-4A might improve the therapeutic efficacy of existing anti-tubercular drugs and provide an attractive strategy for the development of alternative therapies to control tuberculosis.
Collapse
Affiliation(s)
| | - Pampi Chakraborty
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Santosh Kumar
- Tuberculosis Aerosol Challenge Facility, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Prafull Kumar Singh
- Tuberculosis Aerosol Challenge Facility, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - M. G. R. Rajan
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Krishna B. Sainis
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Savita Kulkarni
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
- * E-mail:
| |
Collapse
|
39
|
Choudhary GS, Yao X, Wang J, Peng B, Bader RA, Ren D. Human Granulocyte Macrophage Colony-Stimulating Factor Enhances Antibiotic Susceptibility of Pseudomonas aeruginosa Persister Cells. Sci Rep 2015; 5:17315. [PMID: 26616387 PMCID: PMC4663479 DOI: 10.1038/srep17315] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022] Open
Abstract
Bacterial persister cells are highly tolerant to antibiotics and cause chronic infections. However, little is known about the interaction between host immune systems with this subpopulation of metabolically inactive cells, and direct effects of host immune factors (in the absence of immune cells) on persister cells have not been studied. Here we report that human granulocyte macrophage-colony stimulating factor (GM-CSF) can sensitize the persister cells of Pseudomonas aeruginosa PAO1 and PDO300 to multiple antibiotics including ciprofloxacin, tobramycin, tetracycline, and gentamicin. GM-CSF also sensitized the biofilm cells of P. aeruginosa PAO1 and PDO300 to tobramycin in the presence of biofilm matrix degrading enzymes. The DNA microarray and qPCR results indicated that GM-CSF induced the genes for flagellar motility and pyocin production in the persister cells, but not the normal cells of P. aeruginosa PAO1. Consistently, the supernatants from GM-CSF treated P. aeruginosa PAO1 persister cell suspensions were found cidal to the pyocin sensitive strain P. aeruginosa PAK. Collectively, these findings suggest that host immune factors and bacterial persisters may directly interact, leading to enhanced susceptibility of persister cells to antibiotics.
Collapse
Affiliation(s)
- Geetika S Choudhary
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Xiangyu Yao
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Jing Wang
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Bo Peng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Rebecca A Bader
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Dacheng Ren
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.,Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY 13244, USA.,Department of Civil and Environmental Engineering, Syracuse University, Syracuse, NY 13244, USA.,Department of Biology, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
40
|
Downregulation of the Musca domestica peptidoglycan recognition protein SC (PGRP-SC) leads to overexpression of antimicrobial peptides and tardy pupation. Mol Immunol 2015; 67:465-74. [DOI: 10.1016/j.molimm.2015.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/03/2015] [Accepted: 08/06/2015] [Indexed: 02/05/2023]
|
41
|
Zhang WQ, Huang SH, Huang X, Li JH, Ye P, Xu J, Zheng PZ, Shen HY, Huang JR. Regulation of human mesenchymal stem cell differentiation by TREM-2. Hum Immunol 2015; 77:476-82. [PMID: 26079507 DOI: 10.1016/j.humimm.2015.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 01/31/2015] [Accepted: 06/02/2015] [Indexed: 10/23/2022]
Abstract
Activation of the triggering receptor expressed on myeloid cells 2 (TREM-2) regulates myeloid cell function in vitro. However, the failure to detect TREM-2 protein expression in vivo has hampered studies on immunological and other physiological TREM-2 functions. This study demonstrates that TREM-2 is expressed by human mesenchymal stem cells (h-MSCs) and responds to the toll-like receptor (TLR) ligand lipopolysaccharide (LPS). Knockdown of TREM-2 in h-MSCs using a small interfering RNA (siRNA) reduced the expression levels of TLR2, TLR4, and TLR6, inhibited osteogenic, chondrogenic, and adipogenic differentiation under specific induction conditions, and enhanced LPS-evoked inflammatory cytokine production. Thus, activation of TREM-2 may restrain h-MSC immune activation and promote differentiation for tissue repair.
Collapse
Affiliation(s)
- Wei-Qiong Zhang
- Department of Orthopedics, The Sun Yat Sen Memory Hospital, Sun Yat Sen University, Guangzhou 510120, China; Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China
| | - Sheng-Hui Huang
- Department of Orthopedics, The Sun Yat Sen Memory Hospital, Sun Yat Sen University, Guangzhou 510120, China; Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China
| | - Xi Huang
- Department of Immunology, Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jian-Hua Li
- Physiological Department of Guangzhou Medical University, Guangzhou 510080, China
| | - Pei Ye
- Department of Orthopedics, The Sun Yat Sen Memory Hospital, Sun Yat Sen University, Guangzhou 510120, China; Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China
| | - Jinhuang Xu
- Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China
| | - Pei-Zhong Zheng
- Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Sun Yat Sen Memory Hospital, Sun Yat Sen University, Guangzhou 510120, China
| | - Jian-Rong Huang
- Department of Orthopedics, The Sun Yat Sen Memory Hospital, Sun Yat Sen University, Guangzhou 510120, China; Department of Orthopedics, Zeng Cheng People's Hospital, Guangzhou 511300, China.
| |
Collapse
|
42
|
|
43
|
Chiesa C, Pacifico L, Natale F, Hofer N, Osborn JF, Resch B. Fetal and early neonatal interleukin-6 response. Cytokine 2015; 76:1-12. [PMID: 25890877 DOI: 10.1016/j.cyto.2015.03.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 12/11/2022]
Abstract
In 1998, a systemic fetal cytokine response, defined as a plasma interleukin-6 (IL-6) value above 11 pg/mL, was reported to be a major independent risk factor for the subsequent development of neonatal morbid events even after adjustments for gestational age and other confounders. Since then, the body of literature investigating the use of blood concentrations of IL-6 as a hallmark of the fetal inflammatory response syndrome (FIRS), a diagnostic marker of early-onset neonatal sepsis (EONS) and a risk predictor of white matter injury (WMI), has grown rapidly. In this article, we critically review: IL-6 biological functions; current evidence on the association between IL-6, preterm birth, FIRS and EONS; IL-6 reference intervals and dynamics in the early neonatal period; IL-6 response during the immediate postnatal period and perinatal confounders; accuracy and completeness of IL-6 diagnostic studies for EONS (according to the Standards for Reporting of Diagnostic Accuracy statement); and recent breakthroughs in the association between fetal blood IL-6, EONS, and WMI.
Collapse
Affiliation(s)
- Claudio Chiesa
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy.
| | - Lucia Pacifico
- Department of Pediatrics and Child Neuropsychiatry, Sapienza University of Rome, 00161 Rome, Italy
| | - Fabio Natale
- Department of Pediatrics and Child Neuropsychiatry, Sapienza University of Rome, 00161 Rome, Italy
| | - Nora Hofer
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, AT-8036 Graz, Austria
| | - John F Osborn
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Bernhard Resch
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, AT-8036 Graz, Austria
| |
Collapse
|
44
|
Cho WK, Jang JP, Choi EJ, Jeon YJ, Jung IA, Kim SH, Jung MH, Kim TG, Suh BK. Association of Toll-like receptor 10 polymorphisms with autoimmune thyroid disease in Korean children. Thyroid 2015; 25:250-5. [PMID: 25295614 DOI: 10.1089/thy.2014.0135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The Toll-like receptors (TLRs) are germline-encoded receptors that play an essential role in initiating the immune response against pathogens. In this study, we assess the association of TLR polymorphism with autoimmune thyroid disease (AITD) in Korean children. METHODS We investigated three polymorphisms in the TLR10 gene (rs4129009, rs11096956, and rs10004195) in 85 Korean AITD patients (Graves' disease, [GD]=50, Hashimoto's disease [HD]=35; thyroid-associated ophthalmopathy [TAO]=23, non-TAO=62; male=16, female=69; mean age=13.4±3.1 years) and 279 healthy control subjects. RESULTS In patients with AITD, the frequencies of the TLR10 rs4129009 A allele (odds raio [OR]=3.9, corrected p=0.04) and rs10004195 T allele (OR=2.8, corrected p=0.02) were higher than in the healthy controls, whereas the TLR10 rs4129009 GG genotype (OR=0.3, corrected p=0.04) and rs10004195 AA genotype (OR=0.4, corrected p=0.02) showed lower frequencies. The TLR10 rs11096956 did not show any significant association. These significant associations were also found in the non-thyroid-associated ophthalmopathy (TAO) group, but not in the TAO group. The haplotype (AGT) frequency of TLR10 rs4129009, rs11096956, and rs10004195 was higher in the AITD group than in healthy controls (OR=2.1, corrected p=0.03). CONCLUSIONS Our results suggest that TLR10 polymorphisms may contribute to the pathogenesis of AITD.
Collapse
Affiliation(s)
- Won Kyoung Cho
- 1 Department of Pediatrics, The Catholic University of Korea , Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cervenak J, Doleschall M, Bender B, Mayer B, Schneider Z, Doleschall Z, Zhao Y, Bősze Z, Hammarström L, Oster W, Kacskovics I. NFκB induces overexpression of bovine FcRn: a novel mechanism that further contributes to the enhanced immune response in genetically modified animals carrying extra copies of FcRn. MAbs 2015; 5:860-71. [PMID: 24492342 DOI: 10.4161/mabs.26507] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Among the many functions of the neonatal Fc receptor (FcRn) for IgG, it binds to IgG-opsonized antigen complexes and propagates their traffic into lysosomes where antigen processing occurs. We previously reported that transgenic (Tg) mice and rabbits that carry multiple copies and overexpress FcRn have augmented humoral immune responses. Nuclear factor-kappa B (NFκB) is a critical molecule in the signaling cascade in the immune response. NFκB induces human FcRn expression and our previous in silico analysis suggested NFκB binding sites in the promoter region of the bovine (b) FcRn α-chain gene (FCGRT). Here, we report the identification of three NFκB transcription binding sites in the promoter region of this gene using luciferase reporter gene technology, electromobility shift assay and supershift analysis. Stimulation of primary bovine endothelial cells with the Toll-like receptor-4 ligand lipopolysaccharide (LPS), which mediates its effect via NFκB, resulted in rapid upregulation of the bFcRn expression and a control gene, bovine E-selectin. This rapid bFcRn gene induction was also observed in the spleen of bFcRn Tg mice treated with intraperitoneally injected LPS, analyzed by northern blot analysis. Finally, NFκB-mediated bFcRn upregulation was confirmed at the protein level in macrophages isolated from the bFcRn Tg mice using flow cytometry with a newly developed FcRn specific monoclonal antibody that does not cross-react with the mouse FcRn. We conclude that NFκB regulates bFcRn expression and thus optimizes its functions, e.g., in the professional antigen presenting cells, and contributes to the much augmented humoral immune response in the bFcRn Tg mice.
Collapse
Affiliation(s)
| | - Márton Doleschall
- Molecular Medicine Research Group; Hungarian Academy of Sciences and Semmelweis University; Budapest, Hungary
| | | | - Balázs Mayer
- Department of Dermatology, Venereology, and Dermatooncology; Semmelweis University; Budapest, Hungary
| | - Zita Schneider
- Department of Immunology; Eötvös Loránd University; Budapest, Hungary
| | - Zoltán Doleschall
- Department of Pathogenetics; National Institute of Oncology; Budapest, Hungary
| | - Yaofeng Zhao
- State Key Laboratory of Agrobiotechnology; China Agricultural University; Beijing, China
| | | | - Lennart Hammarström
- Division of Clinical Immunology; Department of Laboratory Medicine; Karolinska Institute at Karolinska University Hospital Huddinge; Stockholm, Sweden
| | | | - Imre Kacskovics
- ImmunoGenes Kft; Budakeszi, Hungary; Department of Immunology; Eötvös Loránd University; Budapest, Hungary
| |
Collapse
|
46
|
Gooshe M, Abdolghaffari AH, Gambuzza ME, Rezaei N. The role of Toll-like receptors in multiple sclerosis and possible targeting for therapeutic purposes. Rev Neurosci 2014; 25:713-39. [PMID: 24914714 DOI: 10.1515/revneuro-2014-0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023]
Abstract
The interaction between the immune and nervous systems suggests invaluable mechanisms for several pathological conditions, especially neurodegenerative disorders. Multiple sclerosis (MS) is a potentially disabling chronic autoimmune disease, characterized by chronic inflammation and neurodegenerative pathology of the central nervous system. Toll-like receptors (TLRs) are an important family of receptors involved in host defense and in recognition of invading pathogens. The role of TLRs in the pathogenesis of autoimmune disorders such as MS is only starting to be uncovered. Recent studies suggest an ameliorative role of TLR3 and a detrimental role of other TLRs in the onset and progression of MS and experimental autoimmune encephalomyelitis, a murine model of MS. Thus, modulating TLRs can represent an innovative immunotherapeutic approach in MS therapy. This article outlines the role of these TLRs in MS, also discussing TLR-targeted agonist or antagonists that could be used in the different stages of the disease.
Collapse
|
47
|
Khalaf JK, Bowen WS, Bazin HG, Ryter KT, Livesay MT, Ward JR, Evans JT, Johnson DA. Characterization of TRIF selectivity in the AGP class of lipid A mimetics: role of secondary lipid chains. Bioorg Med Chem Lett 2014; 25:547-53. [PMID: 25553892 DOI: 10.1016/j.bmcl.2014.12.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
TLR4 agonists that favor TRIF-dependent signaling and the induction of type 1 interferons may have potential as vaccine adjuvants with reduced toxicity. CRX-547 (4), a member of the aminoalkyl glucosaminide 4-phosphate (AGP) class of lipid A mimetics possessing three (R)-3-decanoyloxytetradecanoyl groups and d-relative configuration in the aglycon, selectively reduces MyD88-dependent signaling resulting in TRIF-selective signaling, whereas the corresponding secondary ether lipid 6a containing (R)-3-decyloxytetradecanoyl groups does not. In order to determine which secondary acyl groups are important for the reduction in MyD88-dependent signaling activity of 4, the six possible ester/ether hybrid derivatives of 4 and 6a were synthesized and evaluated for their ability to induce NF-κB in a HEK293 cell reporter assay. An (R)-3-decanoyloxytetradecanoyl group on the 3-position of the d-glucosamine unit was found to be indispensable for maintaining low NF-κB activity irrespective of the substitutions (decyl or decanoyl) on the other two secondary positions. These results suggest that the carbonyl group of the 3-secondary lipid chain may impede homodimerization and/or conformational changes in the TLR4-MD2 complex necessary for MyD88 binding and pro-inflammatory cytokine induction.
Collapse
Affiliation(s)
- Juhienah K Khalaf
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - William S Bowen
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Donald E. Baxter Biomedical Research Building, 570 South Preston Street, Louisville, KY 40202, USA
| | - Hélène G Bazin
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Kendal T Ryter
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Mark T Livesay
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Jon R Ward
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Jay T Evans
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - David A Johnson
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA.
| |
Collapse
|
48
|
Fassl SK, Austermann J, Papantonopoulou O, Riemenschneider M, Xue J, Bertheloot D, Freise N, Spiekermann C, Witten A, Viemann D, Kirschnek S, Stoll M, Latz E, Schultze JL, Roth J, Vogl T. Transcriptome Assessment Reveals a Dominant Role for TLR4 in the Activation of Human Monocytes by the Alarmin MRP8. THE JOURNAL OF IMMUNOLOGY 2014; 194:575-83. [DOI: 10.4049/jimmunol.1401085] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Wang Z, Han X, He N, Chen Z, Brooks CL. Environmental Effect on Surface Immobilized Biological Molecules. J Phys Chem B 2014; 118:12176-85. [DOI: 10.1021/jp508550d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Zunliang Wang
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Si Pai Lou 2, Nanjing 210096, China
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xiaofeng Han
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Si Pai Lou 2, Nanjing 210096, China
| | - Nongyue He
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Si Pai Lou 2, Nanjing 210096, China
| | - Zhan Chen
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles L. Brooks
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
50
|
Wright KT, Giardina C, Vella AT. Therapeutic targeting of the inflammome. Biochem Pharmacol 2014; 92:184-91. [PMID: 25204592 DOI: 10.1016/j.bcp.2014.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Inflammatory responses can vary depending on a myriad of factors including: (1) the initiating stimulus or trigger, (2) the cell types involved in the response, and (3) the specific effector cytokine-chemokine milieus produced. The compilation of these and other factors in a given mechanistic context is sometimes referred to as the "inflammome". Humans and other higher-order mammals have evolved (over time) several discrete inflammomes to counter the effects of pathogens. However, when these inflammomes are induced inappropriately, they drive the development of chronic inflammatory diseases. The vast majority of biological anti-inflammatory treatments currently being developed are focused on the post hoc inhibition of downstream effectors by anti-cytokine monoclonal antibodies and receptor antagonists. This prevailing "end-point treatment" has even directed a new disease classification paradigm, namely a cytokine-based disease classification, as opposed to a traditional diagnosis based on a particular tissue or organ system dysfunction. Although this approach has a number of advantages, it omits the processes that led to the generation of the inflammatory effectors in the first place. In this review, we will expand the cytokine-based disease taxonomy into an inflammome-based taxonomy that includes interventions that subvert a priori cytokine development and can complement post hoc inhibition.
Collapse
Affiliation(s)
- Kyle T Wright
- Department of Immunology, University of Connecticut Health Center, University of Connecticut Heath Center, MC3710 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Charles Giardina
- Department of Molecular & Cell Biology, University of Connecticut, 91 North Eagleville Road, Unit 3125, Storrs, CT 06269-3125, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, University of Connecticut Heath Center, MC3710 263 Farmington Avenue, Farmington, CT 06030, USA.
| |
Collapse
|