1
|
Verma N, Chouhan D, Meghana A, Tiwari V. Heat shock proteins in chronic pain: From molecular chaperones to pain modulators. Neuropharmacology 2025; 266:110263. [PMID: 39667433 DOI: 10.1016/j.neuropharm.2024.110263] [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: 09/22/2024] [Revised: 11/25/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
Chronic pain is the most prevalent and complex clinical disorder,affecting approximately 30% of people globally. Various intricate alterations in nociceptive pathways responsible for chronic pain are linked to long-term tissue damage or injury to the peripheral or central nervous systems. These include remolding in the phenotype of cells and fluctuations in the expression of proteins such as ion channels, neurotransmitters, and receptors. Heat shock proteins are important molecular chaperone proteins in cell responses to stress, including inflammation, neurodegeneration, and pain signaling. They play a key role in activating glial and endothelial cells and in the production of inflammatory mediators and excitatory amino acids in both peripheral and central nervous systems. In particular, they contribute to central sensitization and hyperactivation within the dorsal horn of the spinal cord. The expression of some HSPs plays a remarkable role in upregulating pain response by acting as scavengers of ROS, controlling inflammatory cytokines. Different HSPs act by different mechanisms and several important pathways have been implicated in targeting HSPs for the treatment of neuropathic pain including p38-mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinases (ERKs), brain-derived neurotrophic factors (BDNF). We summarize the role of HSPs in various preclinical and clinical studies and the crosstalk of HSPs with various nociceptors and other pain models. We also highlighted some artificial intelligence tools and machine learning-assisted drug discovery methods for rapid screening of HSPs in various diseases. Focusing on HSPs could lead to the development of new therapeutics that modulate pain responses and enhance our understanding of pain in various pathological conditions and neurological disorders.
Collapse
Affiliation(s)
- Nivedita Verma
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Allani Meghana
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
2
|
Di Luca A, Bennato F, Ianni A, Martino C, Henry M, Meleady P, Martino G. Label-free liquid chromatography mass spectrometry analysis of changes in broiler liver proteins under transport stress. PLoS One 2024; 19:e0311539. [PMID: 39466737 PMCID: PMC11515959 DOI: 10.1371/journal.pone.0311539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
Transportation duration and distance are significant concerns for animal welfare, particularly in the poultry industry. However, limited proteomic studies have investigated the impact of transport duration on poultry welfare. In this study, mass spectrometry based bottom up proteomics was employed to sensitively and impartially profile the liver tissue proteome of chickens, addressing the issue of animal stress and welfare in response to transportation before slaughter. The liver exudates obtained from Ross 508 chickens exposed to either short or long road transportation underwent quantitative label-free LC-MS proteomic profiling. This method identified a total of 1,368 proteins, among which 35 were found to be significantly different (p < 0.05) and capable of distinguishing between short and long road transportation conditions. Specifically, 23 proteins exhibited up-regulation in the non stressed group, while 12 proteins showed up-regulation in the stressed group. The proteins identified in this pilot study encompassed those linked to homeostasis and cellular energetic balance, including heat shock proteins and the 5'-nucleotidase domain-containing family. These results contribute to a deeper understanding of the proteome in broiler liver tissues, shedding light on poultry adaptability to transport stress. Furthermore, the identified proteins present potential as biomarkers, suggesting promising approaches to enhance poultry care and management within the industry.
Collapse
Affiliation(s)
- Alessio Di Luca
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Francesca Bennato
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Andrea Ianni
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Camillo Martino
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Giuseppe Martino
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| |
Collapse
|
3
|
Bai L, Yang M, Wu J, You R, Chen Q, Cheng Y, Qian Z, Yang X, Wang Y, Liu Y. An injectable adhesive hydrogel for photothermal ablation and antitumor immune activation against bacteria-associated oral squamous cell carcinoma. Acta Biomater 2024; 186:229-245. [PMID: 39038749 DOI: 10.1016/j.actbio.2024.07.027] [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: 04/02/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Pathogenic bacteria are closely associated with the occurrence, development and metastasis of oral squamous cell carcinoma (OSCC). Antibacterial therapy has been considered an enhancement strategy to suppress bacteria-associated tumors and promote anti-tumor immune responses. Herein, we developed an injectable adhesive hydrogel, PNIPAM/DL@TIR, for the in situ photothermal ablation and robust stimulation of antitumor immunity against OSCC colonized by Porphyromonas gingivalis (Pg), one of the major oral pathogenic bacteria. PNIPAM/DL@TIR, composed of poly(N-isopropylacrylamide), demethylated lignin, and TAT peptide-conjugated IR820, was prepared using a simple dissolve-dry-swell solvent exchange method. Upon 808 nm laser irradiation, PNIPAM/DL@TIR exerted photothermal effects to ablate Pg-colonized OSCC and generate dual tumor and bacterial antigens. Owing to its large number of catechol groups, PNIPAM/DL@TIR efficiently captured these antigens to form an in situ antigen repository, thereby eliciting robust and durable antitumor immune responses. Proteomic analysis revealed that the captured antigens comprised both tumor neoantigens and bacterial antigens. The catechol groups endowed PNIPAM/DL@TIR with antioxidant activity, which was also conducive to stimulating antitumor immunity. Altogether, this study develops an injectable adhesive hydrogel and provides a combination strategy for treating bacteria-associated OSCC. STATEMENT OF SIGNIFICANCE: In this study, we developed an injectable adhesive hydrogel, PNIPAM/DL@TIR, for in situ photothermal ablation and robust stimulation of antitumor immunity against OSCC colonized by Porphyromonas gingivalis, one of the major oral pathogenic bacteria. PNIPAM/DL@TIR, which consists of poly(N-isopropylacrylamide), demethylated lignin, and TAT peptide-conjugated IR820 exhibited outstanding photothermal performance. Owing to the presence of catechol groups, PNIPAM/DL@TIR has good bioadhesive properties and can capture protein antigens to form in situ antigen repository, thus initiating robust and long-term antitumor immune responses. In addition, PNIPAM/DL@TIR exhibited strong antioxidant activity that is favorable for promoting antitumor immunity. In the mouse model of OSCC with bacterial infection, PNIPAM/DL@TIR not only ablated the primary tumors upon NIR laser irradiation, but also induced tumor and bacterial vaccination in situ to suppress distant tumors and lung metastasis.
Collapse
Affiliation(s)
- Liya Bai
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Meng Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiaxin Wu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Ran You
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Qian Chen
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yuanyuan Cheng
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhanyin Qian
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoying Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yinsong Wang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Yuanyuan Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
4
|
Yang S, Wei S, Wei F. Extracellular vesicles mediated gastric cancer immune response: tumor cell death or immune escape? Cell Death Dis 2024; 15:377. [PMID: 38816455 PMCID: PMC11139918 DOI: 10.1038/s41419-024-06758-8] [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: 12/01/2023] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
Gastric cancer (GC) is a major global health issue, being the fifth most prevalent cancer and the third highest contributor to cancer-related deaths. Although treatment strategies for GC have diversified, the prognosis for advanced GC remains poor. Hence, there is a critical need to explore new directions for GC treatment to enhance diagnosis, treatment, and patient prognosis. Extracellular vesicles (EVs) have emerged as key players in tumor development and progression. Different sources of EVs carry different molecules, resulting in distinct biological functions. For instance, tumor-derived EVs can promote tumor cell proliferation, alter the tumor microenvironment and immune response, while EVs derived from immune cells carry molecules that regulate immune function and possess tumor-killing capabilities. Numerous studies have demonstrated the crucial role of EVs in the development, immune escape, and immune microenvironment remodeling in GC. In this review, we discuss the role of GC-derived EVs in immune microenvironment remodeling and EVs derived from immune cells in GC development. Furthermore, we provide an overview of the potential uses of EVs in immunotherapy for GC.
Collapse
Affiliation(s)
- Shuo Yang
- Department of the Seventh General surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110000̥, Liaoning Province, PR China
| | - Shibo Wei
- Department of the Seventh General surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110000̥, Liaoning Province, PR China.
| | - Fang Wei
- Department of the Seventh General surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110000̥, Liaoning Province, PR China.
| |
Collapse
|
5
|
Farag MR, Alagawany M, Mahdy EAA, El-Hady E, Abou-Zeid SM, Mawed SA, Azzam MM, Crescenzo G, Abo-Elmaaty AMA. Benefits of Chlorella vulgaris against Cadmium Chloride-Induced Hepatic and Renal Toxicities via Restoring the Cellular Redox Homeostasis and Modulating Nrf2 and NF-KB Pathways in Male Rats. Biomedicines 2023; 11:2414. [PMID: 37760855 PMCID: PMC10525457 DOI: 10.3390/biomedicines11092414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
In our life scenarios, we are involuntarily exposed to many heavy metals that are well-distributed in water, food, and air and have adverse health effects on animals and humans. Cadmium (Cd) is one of the most toxic 10 chemicals reported by The World Health Organization (WHO), affecting organ structure and function. In our present study, we use one of the green microalga Chlorella vulgaris (ChV, 500 mg/kg body weight) to investigate the beneficial effects against CdCl2-induced hepato-renal toxicity (Cd, 2 mg/kg body weight for 10 days) on adult male Sprague-Dawley rats. In brief, 40 adult male rats were divided into four groups (n = 10); Control, ChV, Cd, and Cd + ChV. Cadmium alters liver and kidney architecture and disturbs the cellular signaling cascade, resulting in loss of body weight, alteration of the hematological picture, and increased ALT, AST, ALP, and urea in the blood serum. Moreover, cadmium puts hepatic and renal cells under oxidative stress due to the up-regulation of lipid peroxidation resulting in a significant increase in the IgG level as an innate immunity protection and induction of the pro-inflammatory cytokines (IL-1β and TNF-α) that causes hepatic hemorrhage, irregular hepatocytes in the liver and focal glomeruli swelling and proximal tubular degeneration in the kidney. ChV additive to CdCl2, could organize the protein translation process via NF-kB/Nrf2 pathways to prevent oxidative damage by maintaining cellular redox homeostasis and improving the survival of and tolerance of cells against oxidative damage caused by cadmium. The present study shed light on the anti-inflammatory and antioxidative properties of Chlorella vulgaris that suppress the toxicity influence of CdCl2.
Collapse
Affiliation(s)
- Mayada R. Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| | - Eman A. A. Mahdy
- Anatomy and Embryology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt; (E.A.A.M.); (E.E.-H.)
| | - Enas El-Hady
- Anatomy and Embryology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt; (E.A.A.M.); (E.E.-H.)
| | - Shimaa M. Abou-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 6012201, Egypt;
| | - Suzan A. Mawed
- Zoology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt;
| | - Mahmoud M. Azzam
- Department of Animal Production, College of Food & Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Giuseppe Crescenzo
- Department of Veterinary Medicine, University of Bari “Aldo Moro”, Valenzano, 70010 Bari, Italy;
| | - Azza M. A. Abo-Elmaaty
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| |
Collapse
|
6
|
Ghorbaninezhad F, Alemohammad H, Najafzadeh B, Masoumi J, Shadbad MA, Shahpouri M, Saeedi H, Rahbarfarzam O, Baradaran B. Dendritic cell-derived exosomes: A new horizon in personalized cancer immunotherapy? Cancer Lett 2023; 562:216168. [PMID: 37031915 DOI: 10.1016/j.canlet.2023.216168] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
Dendritic cells (DCs) release nanometer-sized membrane vesicles known as dexosomes, containing different molecules, particularly proteins, for presenting antigens, i.e., major histocompatibility complex (MHC)-I/II and CD86. Dexosomes can, directly and indirectly, stimulate antigen-reactive CD8+ and CD4+ T cell responses. Antigen-loaded dexosomes can lead to the development of potent anti-tumoral immune responses. Notably, developing dexosome-based cell-free vaccines could serve as a new vaccination platform in the era of immunotherapy for various cancers. Furthermore, combining dexosomes vaccination strategies with other treatment approaches can considerably increase tumor-specific T cell responses. Herein, we aimed to review how dexosomes interact with immune cells, e.g., CD4+ and CD8+ T cells and natural killer (NK) cells. Besides, we discussed the limitations of this approach and suggested potential strategies to improve its effectiveness for affected patients.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Shahpouri
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbarfarzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Hazrati A, Soudi S, Malekpour K, Mahmoudi M, Rahimi A, Hashemi SM, Varma RS. Immune cells-derived exosomes function as a double-edged sword: role in disease progression and their therapeutic applications. Biomark Res 2022; 10:30. [PMID: 35550636 PMCID: PMC9102350 DOI: 10.1186/s40364-022-00374-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/03/2022] [Indexed: 02/08/2023] Open
Abstract
Exosomes, ranging in size from 30 to 150 nm as identified initially via electron microscopy in 1946, are one of the extracellular vesicles (EVs) produced by many cells and have been the subject of many studies; initially, they were considered as cell wastes with the belief that cells produced exosomes to maintain homeostasis. Nowadays, it has been found that EVs secreted by different cells play a vital role in cellular communication and are usually secreted in both physiological and pathological conditions. Due to the presence of different markers and ligands on the surface of exosomes, they have paracrine, endocrine and autocrine effects in some cases. Immune cells, like other cells, can secrete exosomes that interact with surrounding cells via these vesicles. Immune system cells-derived exosomes (IEXs) induce different responses, such as increasing and decreasing the transcription of various genes and regulating cytokine production. This review deliberate the function of innate and acquired immune cells derived exosomes, their role in the pathogenesis of immune diseases, and their therapeutic appliances.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Kosar Malekpour
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arezou Rahimi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| |
Collapse
|
8
|
The Role of Polyphenols in Regulation of Heat Shock Proteins and Gut Microbiota in Weaning Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6676444. [PMID: 34531940 PMCID: PMC8440081 DOI: 10.1155/2021/6676444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/06/2021] [Indexed: 12/25/2022]
Abstract
Gut microbiota is the natural residents of the intestinal ecosystem which display multiple functions that provide beneficial effects on host physiology. Disturbances in gut microbiota in weaning stress are regulated by the immune system and oxidative stress-related protein pathways. Weaning stress also alters gut microbiota response, limits digestibility, and influences animal productive performance through the production of inflammatory molecules. Heat shock proteins are the molecular chaperones that perform array functions from physiological to pathological point of view and remodeling cellular stress response. As it is involved in the defense mechanism, polyphenols ensure cellular tolerance against enormous stimuli. Polyphenols are nature-blessed compounds that show their existence in plenty of amounts. Due to their wider availability and popularity, they can exert strong immunomodulatory, antioxidative, and anti-inflammatory activities. Their promising health-promoting effects have been demonstrated in different cellular and animal studies. Dietary interventions with polyphenols may alter the gut microbiome response and attenuate the weaning stress related to inflammation. Further, polyphenols elicit health-favored effects through ameliorating inflammatory processes to improve digestibility and thereby exert a protective effect on animal production. Here, in this article, we will expand the role of dietary polyphenol intervention strategies in weaning stress which perturbs gut microbiota function and also paid emphasis to heat shock proteins in gut health. This review article gives new direction to the feed industry to formulate diet containing polyphenols which would have a significant impact on animal health.
Collapse
|
9
|
Jover-Mengual T, Hwang JY, Byun HR, Court-Vazquez BL, Centeno JM, Burguete MC, Zukin RS. The Role of NF-κB Triggered Inflammation in Cerebral Ischemia. Front Cell Neurosci 2021; 15:633610. [PMID: 34040505 PMCID: PMC8141555 DOI: 10.3389/fncel.2021.633610] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral ischemia is a devastating disease that affects many people worldwide every year. The neurodegenerative damage as a consequence of oxygen and energy deprivation, to date, has no known effective treatment. The ischemic insult is followed by an inflammatory response that involves a complex interaction between inflammatory cells and molecules which play a role in the progression towards cell death. However, there is presently a matter of controversy over whether inflammation could either be involved in brain damage or be a necessary part of brain repair. The inflammatory response is triggered by inflammasomes, key multiprotein complexes that promote secretion of pro-inflammatory cytokines. An early event in post-ischemic brain tissue is the release of certain molecules and reactive oxygen species (ROS) from injured neurons which induce the expression of the nuclear factor-kappaB (NF-κB), a transcription factor involved in the activation of the inflammasome. There are conflicting observations related to the role of NF-κB. While some observe that NF-κB plays a damaging role, others suggest it to be neuroprotective in the context of cerebral ischemia, indicating the need for additional investigation. Here we discuss the dual role of the major inflammatory signaling pathways and provide a review of the latest research aiming to clarify the relationship between NF-κB mediated inflammation and neuronal death in cerebral ischemia.
Collapse
Affiliation(s)
- Teresa Jover-Mengual
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, United States
| | - Hyae-Ran Byun
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Brenda L Court-Vazquez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - José M Centeno
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
10
|
Nikfarjam S, Rezaie J, Kashanchi F, Jafari R. Dexosomes as a cell-free vaccine for cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:258. [PMID: 33228747 PMCID: PMC7686678 DOI: 10.1186/s13046-020-01781-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/13/2020] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) secrete vast quantities of exosomes termed as dexosomes. Dexosomes are symmetric nanoscale heat-stable vesicles that consist of a lipid bilayer displaying a characteristic series of lipid and protein molecules. They include tetraspanins and all established proteins for presenting antigenic material such as the major histocompatibility complex class I/II (MHC I/II) and CD1a, b, c, d proteins and CD86 costimulatory molecule. Dexosomes contribute to antigen-specific cellular immune responses by incorporating the MHC proteins with antigen molecules and transferring the antigen-MHC complexes and other associated molecules to naïve DCs. A variety of ex vivo and in vivo studies demonstrated that antigen-loaded dexosomes were able to initiate potent antitumor immunity. Human dexosomes can be easily prepared using monocyte-derived DCs isolated by leukapheresis of peripheral blood and treated ex vivo by cytokines and other factors. The feasibility of implementing dexosomes as therapeutic antitumor vaccines has been verified in two phase I and one phase II clinical trials in malignant melanoma and non small cell lung carcinoma patients. These studies proved the safety of dexosome administration and showed that dexosome vaccines have the capacity to trigger both the adaptive (T lymphocytes) and the innate (natural killer cells) immune cell recalls. In the current review, we will focus on the perspective of utilizing dexosome vaccines in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Sepideh Nikfarjam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, Shafa St, Ershad Blvd., 57147, Urmia, Iran
| | - Fatah Kashanchi
- School of Systems Biology, Laboratory of Molecular Virology, George Mason University, Discovery Hall Room 182, 10900 University Blvd., VA, 20110, Manassas, USA.
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, Shafa St, Ershad Blvd., 57147, Urmia, Iran. .,Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
11
|
Payne M, Bossmann SH, Basel MT. Direct treatment versus indirect: Thermo-ablative and mild hyperthermia effects. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1638. [PMID: 32352660 DOI: 10.1002/wnan.1638] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/02/2020] [Accepted: 04/07/2020] [Indexed: 11/11/2022]
Abstract
Hyperthermia is a rapidly growing field in cancer therapy and many advances have been made in understanding and applying the mechanisms of hyperthermia. Secondary effects of hyperthermia have been increasingly recognized as important in therapeutic effects and multiple studies have started to elucidate their implications for treatment. Immune effects have especially been recognized as important in the efficacy of hyperthermia treatment of cancer. Both thermo-ablative and mild hyperthermia activate the immune system, but mild hyperthermia seems to be more effective at doing so. This may suggest that mild hyperthermia has some advantages over thermo-ablative hyperthermia and research into immune effects of mild hyperthermia should continue. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery.
Collapse
Affiliation(s)
- Macy Payne
- Department of Chemistry, Kansas State University, Manhattan, Kansas, USA
| | - Stefan H Bossmann
- Department of Chemistry, Kansas State University, Manhattan, Kansas, USA
| | - Matthew T Basel
- Department of Anatomy & Physiology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
12
|
Wang C, Huang X, Wu Y, Wang J, Li F, Guo G. Tumor Cell-associated Exosomes Robustly Elicit Anti-tumor Immune Responses through Modulating Dendritic Cell Vaccines in Lung Tumor. Int J Biol Sci 2020; 16:633-643. [PMID: 32025211 PMCID: PMC6990923 DOI: 10.7150/ijbs.38414] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 12/14/2019] [Indexed: 12/15/2022] Open
Abstract
DC vaccine-based immunotherapy is emerging as a novel therapeutic strategy for cancer treatment, however, antitumor effect of DC vaccines based on tumor cell lysates (TCLs) remains unsatisfactory due to poor immunogenicity of tumor antigens. Although tumor-associated exosomes (TAEs) have been reported as a promising antigen for DC vaccines, it remains unclear how TAE-based DC vaccine induced antitumor immunity in lung cancer. Methods: In the present study, we extracted TAEs from the supernatant of tumor cell culture medium, and compared the effect of TAEs with TCLs on DCs. To further evaluate the therapeutic effect of DCTAE, we used immunofluorescence and flow cytometry to evaluate the apoptosis of tumor tissue, tumor-infiltrating CD8+ T cells and Tregs in TDLNs and spleen. Then the levels of cytokines of IL-12, IFN-γ, L-10 and TGF-β were quantified by ELISA assays. Results: Our data showed that TAEs were more potent than TCLs to promote DC maturation and enhance MHC cross presentation, which directly contributed to more robust tumor-specific cytotoxic T lymphocyte (CTL) response. More importantly, TAEs reduced the expression of PD-L1 of DCs, thereby led to down-regulated population of Tregs in vitro. Moreover, DCTAE remarkably suppressed the tumor growth and prolonged survival rate in vivo, due to participance of CD8+ T cells and decreased Tregs in TDLNs and spleen. Conclusion: TAEs could serve to improve vaccine-elicited immunotherapy by triggering stronger DC-mediated immune responses and decreasing Tregs in the tumor microenvironment.
Collapse
Affiliation(s)
- Ce Wang
- Department of anatomy, School of Medicine, Jinan University, Guangzhou 510632, China.,Shenzhen Key Laboratory of Stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Xue Huang
- Shenzhen Key Laboratory of Stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Yingjuan Wu
- Shenzhen Key Laboratory of Stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Jingbo Wang
- Shenzhen Key Laboratory of Stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Furong Li
- Shenzhen Key Laboratory of Stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Guoqing Guo
- Department of anatomy, School of Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
13
|
Ali SE, Waddington JC, Park BK, Meng X. Definition of the Chemical and Immunological Signals Involved in Drug-Induced Liver Injury. Chem Res Toxicol 2019; 33:61-76. [PMID: 31682113 DOI: 10.1021/acs.chemrestox.9b00275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Idiosyncratic drug-induced liver injury (iDILI), which is rare and often recognized only late in drug development, poses a major public health concern and impediment to drug development due to its high rate of morbidity and mortality. The mechanisms of DILI are not completely understood; both non-immune- and immune-mediated mechanisms have been proposed. Non-immune-mediated mechanisms including direct damage to hepatocytes, mitochondrial toxicity, interference with transporters, and alteration of bile ducts are well-known to be associated with drugs such as acetaminophen and diclofenac; whereas immune-mediated mechanisms involving activation of both adaptive and innate immune cells and the interactions of these cells with parenchymal cells have been proposed. The chemical signals involved in activation of both innate and adaptive immune responses are discussed with respect to recent scientific advances. In addition, the immunological signals including cytokine and chemokines that are involved in promoting liver injury are also reviewed. Finally, we discuss how liver tolerance and regeneration can have profound impact on the pathogenesis of iDILI. Continuous research in developing in vitro systems incorporating immune cells with liver cells and animal models with impaired liver tolerance will provide an opportunity for improved prediction and prevention of immune-mediated iDILI.
Collapse
Affiliation(s)
- Serat-E Ali
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - James C Waddington
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| |
Collapse
|
14
|
Besztercei B, Vancsik T, Benedek A, Major E, Thomas MJ, Schvarcz CA, Krenács T, Benyó Z, Balogh A. Stress-Induced, p53-Mediated Tumor Growth Inhibition of Melanoma by Modulated Electrohyperthermia in Mouse Models without Major Immunogenic Effects. Int J Mol Sci 2019; 20:ijms20164019. [PMID: 31426515 PMCID: PMC6720184 DOI: 10.3390/ijms20164019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Modulated electrohyperthermia (mEHT), an innovative complementary technique of radio-, chemo-, and targeted oncotherapy modalities, can induce tumor apoptosis and contribute to a secondary immune-mediated cancer death. Here, we tested the efficiency of high-fever range (~42 °C) mEHT on B16F10 melanoma both in cell culture and allograft models. In vivo, mEHT treatment resulted in significant tumor size reduction when repeated three times, and induced major stress response as indicated by upregulated cytoplasmic and cell membrane hsp70 levels. Despite the increased PUMA and apoptosis-inducing factor 1, and moderate rise in activated-caspase-3, apoptosis was not significant. However, phospho-H2AX indicated DNA double-strand breaks, which upregulated p53 protein and its downstream cyclin-dependent kinase inhibitors p21waf1 and p27kip. Combined in vitro treatment with mEHT and the p53 activator nutlin-3a additively reduced cell viability compared to monotherapies. Though mEHT promoted the release of damage-associated molecular pattern (DAMP) damage signaling molecules hsp70, HMGB1 and ATP to potentiate the tumor immunogenicity of melanoma allografts, it reduced MHC-I and melan-A levels in tumor cells. This might explain why the number of cytotoxic T cells was moderately reduced, while the amount of natural killer (NK) cells was mainly unchanged and only macrophages increased significantly. Our results suggest that mEHT-treatment-related tumor growth control was primarily mediated by cell-stress-induced p53, which upregulated cyclin-dependent kinase inhibitors. The downregulated tumor antigen-presenting machinery may explain the reduced cytotoxic T-cell response despite increased DAMP signaling. Decreased tumor antigen and MHC-I levels suggest that natural killer (NK) cells and macrophages were the major contributors to tumor eradication.
Collapse
Affiliation(s)
- Balázs Besztercei
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Tamás Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1097 Budapest, Hungary
| | - Anett Benedek
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Enikő Major
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Mbuotidem J Thomas
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Csaba A Schvarcz
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1097 Budapest, Hungary
| | - Zoltán Benyó
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary
| | - Andrea Balogh
- Institute of Clinical Experimental Research, Semmelweis University, 1097 Budapest, Hungary.
| |
Collapse
|
15
|
Farag MR, Elhady WM, Ahmed SYA, Taha HSA, Alagawany M. Astragalus polysaccharides alleviate tilmicosin-induced toxicity in rats by inhibiting oxidative damage and modulating the expressions of HSP70, NF-kB and Nrf2/HO-1 pathway. Res Vet Sci 2019; 124:137-148. [PMID: 30901666 DOI: 10.1016/j.rvsc.2019.03.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/02/2019] [Accepted: 03/12/2019] [Indexed: 12/22/2022]
Abstract
The present study evaluated the toxic effects of Tilmicosin (TIL) on adult rats. The rats received a single subcutaneous injection of TIL at different doses (10, 25, 50, 75 and 100 mg/kg bw). TIL altered the biochemical parameters including liver and kidney function markers, glucose level and lipid profile as well as resulted in histopathological lesions in liver and adrenal glands mostly in rats exposed to 75 and 100 mg/kg bw. Then the role of Astragalus polysaccharide (APS) at 100 and 200 mg/kg bw, in modulating the toxic effects induced by high dose of TIL was evaluated. Single injection of TIL at a dose of 75 mg/kg bw was found to increase the activity of ALT, AST and ALP enzymes, induce the generation of reactive oxygen species (ROS) and decrease the total antioxidant capacity (TAC). TIL upregulated the hepatic mRNA expression of heat shock protein 70 (HSP70) and nuclear factor kappa B (NF-kB) while blocked the Nrf2/HO-1 mediated response. These changes were also associated with increasing tumer necrosis factor-alpha (TNF-α), interlukin1-beta (IL-1β) and nitric oxide levels. On the other hand, the results indicate that APS has a beneficial role particularly at high level in alleviating the stress and the hepatotoxic effects elicited by TIL injection in rats.
Collapse
Affiliation(s)
- Mayada Ragab Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt.
| | - Wlaa M Elhady
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt
| | - Sarah Y A Ahmed
- Microbiology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt
| | - Heba S A Taha
- Genetic Department, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
16
|
Liu Y, Ou Y, Sun L, Li W, Yang J, Zhang X, Hu Y. Alcohol dehydrogenase of Candida albicans triggers differentiation of THP-1 cells into macrophages. J Adv Res 2019; 18:137-145. [PMID: 30923636 PMCID: PMC6424053 DOI: 10.1016/j.jare.2019.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 12/21/2022] Open
Abstract
Candida albicans proteins located on the cell wall and in the cytoplasm have gained great attention because they are not only involved in cellular metabolism and the maintenance of integrity but also interact with host immune systems. Previous research has reported that enolase from C. albicans exhibits high immunogenicity and effectively protects mice against disseminated candidiasis. In this study, alcohol dehydrogenase (ADH) of C. albicans was cloned and purified for the first time, and this study focused on evaluating its effects on the differentiation of the human monocytic cell line THP-1. The morphological features of THP-1 cells exposed to ADH were similar to those of phorbol-12-myristate acetate-differentiated (PMA-differentiated) macrophages. Functionally, ADH enhanced the adhesion, phagocytosis, and killing capacities of THP-1 cells. A flow cytometric assay demonstrated that ADH-induced THP-1 cells significantly increased CD86 and CD11b expression. The production of IL-1β, IL-6, and TNF-α by cells increased in the presence of ADH. As expected, after pretreatment with a MEK inhibitor (U0126), ADH-induced THP-1 cells exhibited unaltered morphological features, eliminated ERK1/2 phosphorylation, prevented CD86/CD11b upregulation and inhibited pro-inflammatory cytokine increase. Collectively, these results suggest that ADH enables THP-1 cells to differentiate into macrophages via the ERK pathway, and it may play an important role in the immune response against fungal invasion.
Collapse
Affiliation(s)
- Yanglan Liu
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China
| | - Yuxue Ou
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China
| | - Luping Sun
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China
| | - Wenqing Li
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China
| | - Jinghong Yang
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China
| | - Xiaohuan Zhang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yan Hu
- Department of Oral Biology, School of Stomatology, Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Lee S, Son B, Park G, Kim H, Kang H, Jeon J, Youn H, Youn B. Immunogenic Effect of Hyperthermia on Enhancing Radiotherapeutic Efficacy. Int J Mol Sci 2018; 19:E2795. [PMID: 30227629 PMCID: PMC6164993 DOI: 10.3390/ijms19092795] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022] Open
Abstract
Hyperthermia is a cancer treatment where tumor tissue is heated to around 40 °C. Hyperthermia shows both cancer cell cytotoxicity and immune response stimulation via immune cell activation. Immunogenic responses encompass the innate and adaptive immune systems, involving the activation of macrophages, natural killer cells, dendritic cells, and T cells. Moreover, hyperthermia is commonly used in combination with different treatment modalities, such as radiotherapy and chemotherapy, for better clinical outcomes. In this review, we will focus on hyperthermia-induced immunogenic effects and molecular events to improve radiotherapy efficacy. The beneficial potential of integrating radiotherapy with hyperthermia is also discussed.
Collapse
Affiliation(s)
- Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Gaeul Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Jaewan Jeon
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea.
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
18
|
Caruso Bavisotto C, Graziano F, Rappa F, Marino Gammazza A, Logozzi M, Fais S, Maugeri R, Bucchieri F, Conway de Macario E, Macario AJL, Cappello F, Iacopino DG, Campanella C. Exosomal Chaperones and miRNAs in Gliomagenesis: State-of-Art and Theranostics Perspectives. Int J Mol Sci 2018; 19:E2626. [PMID: 30189598 PMCID: PMC6164348 DOI: 10.3390/ijms19092626] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/19/2022] Open
Abstract
Gliomas have poor prognosis no matter the treatment applied, remaining an unmet clinical need. As background for a substantial change in this situation, this review will focus on the following points: (i) the steady progress in establishing the role of molecular chaperones in carcinogenesis; (ii) the recent advances in the knowledge of miRNAs in regulating gene expression, including genes involved in carcinogenesis and genes encoding chaperones; and (iii) the findings about exosomes and their cargo released by tumor cells. We would like to trigger a discussion about the involvement of exosomal chaperones and miRNAs in gliomagenesis. Chaperones may be either targets for therapy, due to their tumor-promoting activity, or therapeutic agents, due to their antitumor growth activity. Thus, chaperones may well represent a Janus-faced approach against tumors. This review focuses on extracellular chaperones as part of exosomes' cargo, because of their potential as a new tool for the diagnosis and management of gliomas. Moreover, since exosomes transport chaperones and miRNAs (the latter possibly related to chaperone gene expression in the recipient cell), and probably deliver their cargo in the recipient cells, a new area of investigation is now open, which is bound to generate significant advances in the understanding and treatment of gliomas.
Collapse
Affiliation(s)
- Celeste Caruso Bavisotto
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
- Institute of Biophysics, National Research Council, 90143 Palermo, Italy.
| | - Francesca Graziano
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Francesca Rappa
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Antonella Marino Gammazza
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Rosario Maugeri
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Alberto J L Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA.
| | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| | - Domenico G Iacopino
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Neurosurgery, University of Palermo, 90127 Palermo, Italy.
| | - Claudia Campanella
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy.
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90136 Palermo, Italy.
| |
Collapse
|
19
|
Romero-López JP, Domínguez-López ML, Burgos-Vargas R, García-Latorre E. Stress proteins in the pathogenesis of spondyloarthritis. Rheumatol Int 2018; 39:595-604. [PMID: 29855675 DOI: 10.1007/s00296-018-4070-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/26/2018] [Indexed: 12/15/2022]
Abstract
Spondyloarthritis is an autoinflammatory rheumatic disease in which arthritis and osteoproliferation lead the patients who suffer from it to chronic disability. This disease is associated with the expression of class I MHC molecule HLA-B27, which tends to be misfolded in the endoplasmic reticulum and, therefore, expressed in aberrant forms. This phenomena lead to endoplasmic reticulum stress, which in time, evokes a whole response to cellular injury. Under these conditions, the molecules involved in restoring cell homeostasis play a key role. Such is the case of the "heat-shock proteins", which usually regulate protein folding, but also have important immunomodulatory functions, as well as some roles in tissue modeling. In this review, we attempt to summarize the involvement of cell stress and heat-shock proteins in the homeostatic disturbances and pathological conditions associated with this disease.
Collapse
Affiliation(s)
- José Pablo Romero-López
- Laboratorio de Inmunoquímica I, Departmento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala SN, CP 11340, Ciudad de México, México
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica I, Departmento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala SN, CP 11340, Ciudad de México, México
| | - Rubén Burgos-Vargas
- Departamento de Reumatología, Hospital General de México "Dr. Eduardo Liceaga", Ciudad de México, México
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica I, Departmento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala SN, CP 11340, Ciudad de México, México.
| |
Collapse
|
20
|
Menga M, Trotta R, Scrima R, Pacelli C, Silvestri V, Piccoli C, Capitanio N, Liso A. Febrile temperature reprograms by redox-mediated signaling the mitochondrial metabolic phenotype in monocyte-derived dendritic cells. Biochim Biophys Acta Mol Basis Dis 2017; 1864:685-699. [PMID: 29246446 DOI: 10.1016/j.bbadis.2017.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/15/2017] [Accepted: 12/08/2017] [Indexed: 02/06/2023]
Abstract
Fever-like hyperthermia is known to stimulate innate and adaptive immune responses. Hyperthermia-induced immune stimulation is also accompanied with, and likely conditioned by, changes in the cell metabolism and, in particular, mitochondrial metabolism is now recognized to play a pivotal role in this context, both as energy supplier and as signaling platform. In this study we asked if challenging human monocyte-derived dendritic cells with a relatively short-time thermal shock in the fever-range, typically observed in humans, caused alterations in the mitochondrial oxidative metabolism. We found that following hyperthermic stress (3h exposure at 39°C) TNF-α-releasing dendritic cells undergo rewiring of the oxidative metabolism hallmarked by decrease of the mitochondrial respiratory activity and of the oxidative phosphorylation and increase of lactate production. Moreover, enhanced production of reactive oxygen and nitrogen species and accumulation of mitochondrial Ca2+ was consistently observed in hyperthermia-conditioned dendritic cells and exhibited a reciprocal interplay. The hyperthermia-induced impairment of the mitochondrial respiratory activity was (i) irreversible following re-conditioning of cells to normothermia, (ii) mimicked by exposing normothermic cells to the conditioned medium of the hyperthermia-challenged cells, (iii) largely prevented by antioxidant and inhibitors of the nitric oxide synthase and of the mitochondrial calcium porter, which also inhibited release of TNF-α. These observations combined with gene expression analysis support a model based on a thermally induced autocrine signaling, which rewires and sets a metabolism checkpoint linked to immune activation of dendritic cells.
Collapse
Affiliation(s)
- Marta Menga
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Rosa Trotta
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Veronica Silvestri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
21
|
Abstract
As potent antigen-presenting cells, dendritic cells (DCs) comprise the most heterogeneous cell population with significant cellular phenotypic and functional plasticity. They form a sentinel network to modulate immune responses, since intrinsic cellular mechanisms and complex external, environmental signals endow DCs with the distinct capacity to induce protective immunity or tolerance to self. Interactions between DCs and other cells of the immune system mediate this response. This interactive response depends on DC maturation status and subtype, as well as the microenvironment of the tissue location and DC-intrinsic regulators. Dysregulated DCs can initiate and perpetuate various immune disorders, which creates attractive therapeutic targets. In this review, we provide a detailed outlook on DC ontogeny and functional specialization. We highlight recent advances on the regulatory role that DCs play in immune responses, the putative molecular regulators that control DC functional responding and the contribution of DCs to inflammatory disease physiopathology.
Collapse
|
22
|
Ogese MO, Faulkner L, Jenkins RE, French NS, Copple IM, Antoine DJ, Elmasry M, Malik H, Goldring CE, Park BK, Betts CJ, Naisbitt DJ. Characterization of Drug-Specific Signaling Between Primary Human Hepatocytes and Immune Cells. Toxicol Sci 2017; 158:76-89. [DOI: 10.1093/toxsci/kfx069] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
23
|
Rao Q, Zuo B, Lu Z, Gao X, You A, Wu C, Du Z, Yin H. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology 2016; 64:456-472. [PMID: 26990897 DOI: 10.1002/hep.28549] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/04/2016] [Accepted: 03/06/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) remains a global challenge due to high morbidity and mortality rates and poor response to treatment. Immunotherapy, based on introduction of dendritic cells (DCs) activated by tumor cell lysates as antigens ex vivo, shows limited response rates in HCC patients. Here, we demonstrate that tumor cell-derived exosomes (TEXs), displaying an array of HCC antigens, can elicit a stronger immune response than cell lysates in vitro and in vivo. Significant tumor growth inhibition was achieved in ectopic and orthotopic HCC mice treated with TEX-pulsed DCs. Importantly, the tumor immune microenvironment was significantly improved in orthotopic HCC mice treated by TEX-pulsed DCs, demonstrated by increased numbers of T lymphocytes, elevated levels of interferon-γ, and decreased levels of interleukin-10 and tumor growth factor-β in tumor sites. As expected, T cells played an essential role in the TEX-pulsed DC-mediated immune response. Notably, exosomes from HCC cells not only promoted HCC-specific cytolysis but also provided cross-protective effects against pancreatic cancer cells. Moreover, HCC-specific cytolysis, elicited by DCs pulsed with human HepG2 cell-derived exosomes, was observed across different human HCC cells irrespective of human leukocyte antigen types. CONCLUSION HCC TEXs can potently carry HCC antigens, trigger a strong DC-mediated immune response, and improve the HCC tumor microenvironment. (Hepatology 2016;64:456-472).
Collapse
Affiliation(s)
- Quan Rao
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
| | - Bingfeng Zuo
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Zhen Lu
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Xianjun Gao
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Abin You
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
- Tianjin Cancer Hospital, Tianjin Medical University, Hexi District, Tianjin, China
| | - Chenxuan Wu
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
| | - Zhi Du
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
- Department of Hepatobiliary Surgery, Key Laboratory of Artificial Cell, Institute for Hepatobiliary Diseases, Third Central Hospital, Tianjin Medical University, Hedong District, Tianjin, China
| | - HaiFang Yin
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| |
Collapse
|
24
|
Pitt JM, André F, Amigorena S, Soria JC, Eggermont A, Kroemer G, Zitvogel L. Dendritic cell-derived exosomes for cancer therapy. J Clin Invest 2016; 126:1224-32. [PMID: 27035813 DOI: 10.1172/jci81137] [Citation(s) in RCA: 464] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
DC-derived exosomes (Dex) are nanometer-sized membrane vesicles that are secreted by the sentinel antigen-presenting cells of the immune system: DCs. Like DCs, the molecular composition of Dex includes surface expression of functional MHC-peptide complexes, costimulatory molecules, and other components that interact with immune cells. Dex have the potential to facilitate immune cell-dependent tumor rejection and have distinct advantages over cell-based immunotherapies involving DCs. Accordingly, Dex-based phase I and II clinical trials have been conducted in advanced malignancies, showing the feasibility and safety of the approach, as well as the propensity of these nanovesicles to mediate T and NK cell-based immune responses in patients. This Review will evaluate the interactions of Dex with immune cells, their clinical progress, and the future of Dex immunotherapy for cancer.
Collapse
|
25
|
Wick C. Tolerization against atherosclerosis using heat shock protein 60. Cell Stress Chaperones 2016; 21:201-11. [PMID: 26577462 PMCID: PMC4786533 DOI: 10.1007/s12192-015-0659-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 01/06/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the artery wall, and both innate and adaptive immunity play important roles in the pathogenesis of this disease. In several experimental and human experiments of early atherosclerotic lesions, it has been shown that the first pathogenic event in atherogenesis is intimal infiltration of T cells at predilection sites. These T cells react to heat shock protein 60 (HSP60), which is a ubiquitous self-antigen expressed on the surface of endothelial cells (ECs) together with adhesion molecules in response to classical risk factors for atherosclerosis. When HSP60 is expressed on the EC surface, it can act as a "danger-signal" for both cellular and humoral immune reactions. Acquired by infection or vaccination, beneficial protective immunity to microbial HSP60 and bona fide autoimmunity to biochemically altered autologous HSP60 is present in all humans. Thus, the development of atherosclerosis during aging is paid by the price for lifelong protective preexisting anti-HSP60 immunity by harmful (auto)immune cross-reactive attack on arterial ECs maltreated by atherosclerosis risk factors. This is supported by experiments, which shows that bacterial HSP60 immunization can lead and accelerate experimental atherosclerosis. This review article presents accumulating proof that supports the idea that tolerization with antigenic HSP60 protein or its peptides may arrest or even prevent atherosclerosis by increased production of regulatory T cells and/or anti-inflammatory cytokines. Recent data indicates that HSP60, or more likely some of its derivative peptides, has immunoregulatory functions. Therefore, these peptides may have important potential for being used as diagnostic agents or therapeutic targets.
Collapse
Affiliation(s)
- Cecilia Wick
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Center for Molecular Medicine (CMM) L8:04, Karolinska University Hospital Solna, S-17176, Stockholm, Sweden.
- Laboratory of Autoimmunity, Division for Experimental Pathophysiology and Immunology, Biocenter, Innsbruck Medical University, Innsbruck, A-6020, Austria.
| |
Collapse
|
26
|
Arnal ME, Lallès JP. Gut epithelial inducible heat-shock proteins and their modulation by diet and the microbiota. Nutr Rev 2016; 74:181-97. [PMID: 26883882 DOI: 10.1093/nutrit/nuv104] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epidemic of metabolic diseases has raised questions about the interplay between the human diet and the gut and its microbiota. The gut has two vital roles: nutrient absorption and intestinal barrier function. Gut barrier defects are involved in many diseases. Excess energy intake disturbs the gut microbiota and favors body entry of microbial compounds that stimulate chronic metabolic inflammation. In this context, the natural defense mechanisms of gut epithelial cells and the potential to boost them nutritionally warrant further study. One such important defense system is the activation of inducible heat-shock proteins (iHSPs) which protect the gut epithelium against oxidative stress and inflammation. Importantly, various microbial components can induce the expression of iHSPs. This review examines gut epithelial iHSPs as the main targets of microbial signals and nutrients and presents data on diseases involving disturbances of gut epithelial iHSPs. In addition, a broad literature analysis of dietary modulation of gut epithelial iHSPs is provided. Future research aims should include the identification of gut microbes that can optimize gut-protective iHSPs and the evaluation of iHSP-mediated health benefits of nutrients and food components.
Collapse
Affiliation(s)
- Marie-Edith Arnal
- M.E. Arnal and J.P. Lallès are with the Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France. J.P. Lallès is with the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France
| | - Jean-Paul Lallès
- M.E. Arnal and J.P. Lallès are with the Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France. J.P. Lallès is with the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France.
| |
Collapse
|
27
|
The Dichotomy of Tumor Exosomes (TEX) in Cancer Immunity: Is It All in the ConTEXt? Vaccines (Basel) 2015; 3:1019-51. [PMID: 26694473 PMCID: PMC4693230 DOI: 10.3390/vaccines3041019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/24/2015] [Accepted: 12/05/2015] [Indexed: 02/06/2023] Open
Abstract
Exosomes are virus-sized nanoparticles (30–130 nm) formed intracellularly as intravesicular bodies/intralumenal vesicles within maturing endosomes (“multivesicular bodies”, MVBs). If MVBs fuse with the cell’s plasma membrane, the interior vesicles may be released extracellularly, and are termed “exosomes”. The protein cargo of exosomes consists of cytosolic, membrane, and extracellular proteins, along with membrane-derived lipids, and an extraordinary variety of nucleic acids. As such, exosomes reflect the status and identity of the parent cell, and are considered as tiny cellular surrogates. Because of this closely entwined relationship between exosome content and the source/status of the parental cell, conceivably exosomes could be used as vaccines against various pathologies, as they contain antigens associated with a given disease, e.g., cancer. Tumor-derived exosomes (TEX) have been shown to be potent anticancer vaccines in animal models, driving antigen-specific T and B cell responses, but much recent literature concerning TEX strongly places the vesicles as powerfully immunosuppressive. This dichotomy suggests that the context in which the immune system encounters TEX is critical in determining immune stimulation versus immunosuppression. Here, we review literature on both sides of this immune coin, and suggest that it may be time to revisit the concept of TEX as anticancer vaccines in clinical settings.
Collapse
|
28
|
Tanaka T, Kajiwara T, Kutomi G, Kurotaki T, Saito K, Kanaseki T, Tsukahara T, Hirohashi Y, Torigoe T, Hirata K, Okamoto Y, Sato N, Tamura Y. CpG-A stimulates Hsp72 secretion from plasmacytoid dendritic cells, facilitating cross-presentation. Immunol Lett 2015; 167:34-40. [PMID: 26141624 DOI: 10.1016/j.imlet.2015.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 11/19/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are the main producers of IFN-α in response to unmethylated DNA molecules, including cytosine guanine dinucleotide (CpG)-DNA in vivo. pDCs specifically express toll-like receptor (TLR) 9 and are therefore able to recognize the unmethylated DNAs. It has recently been shown that not only conventional DCs (cDCs) but also pDCs efficiently cross-present exogenous antigens after TLR9 activation. However, the precise molecular mechanism has remained unclear. Here, we show that pDCs secreted heat shock protein 72 (Hsp72) in response to CpG-A administration in a TLR9-dependent manner. Extracellular Hsp72 bound to an Hsp90-peptide complex and enhanced binding of Hsp90-peptide complex to pDC, resulting in efficient cross-presentation. Our experiments therefore suggest a mechanism for orchestration of immune responses by stimulation of pDCs with CpG-A.
Collapse
Affiliation(s)
- Tsutomu Tanaka
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan; United Graduate School of Veterinary Sciences, Yamaguchi University, Yamaguchi, Japan
| | - Toshimitsu Kajiwara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Goro Kutomi
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takehiro Kurotaki
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keita Saito
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koichi Hirata
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshiharu Okamoto
- Joint Department of Veterinary Medicine, Tottori University, Tottori, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuaki Tamura
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Molecular Therapeutics, Center for Food & Medical Innovation, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
29
|
Knight M, Elhelu O, Smith M, Haugen B, Miller A, Raghavan N, Wellman C, Cousin C, Dixon F, Mann V, Rinaldi G, Ittiprasert W, Brindley PJ. Susceptibility of Snails to Infection with Schistosomes is influenced by Temperature and Expression of Heat Shock Proteins. ACTA ACUST UNITED AC 2015; 5. [PMID: 26504668 PMCID: PMC4618387 DOI: 10.4172/2161-1165.1000189] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The freshwater snail, Biomphalaria glabrata is the obligate intermediate host for the transmission of the parasitic trematode, Schistosoma mansoni the causative agent of the chronic debilitating neglected tropical disease, schistosomiasis. We showed previously that in juvenile snails, early and significant induction of stress manifested by the expression of stress proteins, Hsp 70, Hsp 90 and reverse transcriptase (RT) of the non- LTR retrotransposon, nimbus, is a characteristic feature of juvenile susceptible NMRI but not resistant BS-90 snails. These latter, however, could be rendered susceptible after mild heat shock at 32°C, revealing that resistance in the BS-90 resistant snail to schistosomes is a temperature dependent trait. Here we tested the hypothesis that maintenance of BS-90 resistant snails at the permissive temperature for several generations affects the resistance phenotype displayed at the non-permissive temperature of 25°C. The progeny of BS-90 snails bred and maintained through several generations (F1 to F4) at 32°C were susceptible to the schistosome infection when returned to room temperature, shedding cercariae at four weeks post-infection. Moreover, the study of expression levels of the heat shock protein (Hsp) 70 protein by ELISA and western blot analysis, showed that this protein is also differentially expressed between susceptible and resistant snails, with susceptible snails expressing more protein than their resistant counterparts after early exposure to wild-type but not to radiation-attenuated miracidia. These data suggested that in the face of global warming, the ability to sustain a reduction in schistosomiasis by using refractory snails as a strategy to block transmission of the disease might prove challenging since non-lethal elevation in temperature, affects snail susceptibility to S. mansoni.
Collapse
Affiliation(s)
- Matty Knight
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA ; George Washington University, Department of Microbiology, School of Medicine and Health Sciences, 2300 Eye Street NW, Washington, D.C, USA
| | - O Elhelu
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - M Smith
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - B Haugen
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - A Miller
- Schistosomiasis Resource Center, Biomedical Research Institute, 12111 Parklawn Drive, Rockville, USA
| | - N Raghavan
- Schistosomiasis Resource Center, Biomedical Research Institute, 12111 Parklawn Drive, Rockville, USA
| | - C Wellman
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - C Cousin
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - F Dixon
- University of the District of Columbia, 4200 Connecticut Ave, Washington, D.C. 20008, USA
| | - V Mann
- George Washington University, Department of Microbiology, School of Medicine and Health Sciences, 2300 Eye Street NW, Washington, D.C, USA
| | - G Rinaldi
- George Washington University, Department of Microbiology, School of Medicine and Health Sciences, 2300 Eye Street NW, Washington, D.C, USA
| | - W Ittiprasert
- Schistosomiasis Resource Center, Biomedical Research Institute, 12111 Parklawn Drive, Rockville, USA
| | - P J Brindley
- George Washington University, Department of Microbiology, School of Medicine and Health Sciences, 2300 Eye Street NW, Washington, D.C, USA
| |
Collapse
|
30
|
Wang J, Wang Z, Mo Y, Zeng Z, Wei P, Li T. Effect of hyperthermic CO 2-treated dendritic cell-derived exosomes on the human gastric cancer AGS cell line. Oncol Lett 2015; 10:71-76. [PMID: 26170979 DOI: 10.3892/ol.2015.3155] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 03/19/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to determine the antitumor effects of hyperthermic CO2 (HT-CO2)-treated dendritic cell (DC)-derived exosomes (Dex) on human gastric cancer AGS cells. Mouse-derived DCs were incubated in HT-CO2 at 43°C for 4 h. The exosomes in the cell culture supernatant were then isolated. Cell proliferation was analyzed using the cell counting kit-8 (CCK-8) assay. Cell apoptosis was observed using flow cytometry, Hoechst 33258 staining and the analysis of caspase-3 activity. In addition, the proliferation of tumor cells was evaluated in xenotransplant nude mice. HT-CO2 markedly inhibited cell proliferation, as assessed by the CCK-8 assay, and also induced apoptosis in a time-dependent manner, as demonstrated by Annexin V/propidium iodide flow cytometry, caspase-3 activity and morphological analysis using Hoechst fluorescent dye. It was also revealed that HT-CO2-treated Dex decreased the expression of heat shock protein 70 and inhibited tumor growth in nude mice. In conclusion, HT-CO2 exerted an efficacious immune-enhancing effect on DCs. These findings may provide a novel strategy for the elimination of free cancer cells during laparoscopic resection. However, the potential cellular mechanisms underlying this process require further investigation.
Collapse
Affiliation(s)
- Jinlin Wang
- Department of General Surgery, Dongguan People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Zhiyong Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| | - Yanxia Mo
- Department of General Surgery, Dongguan People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Zhaohui Zeng
- Department of General Surgery, Dongguan People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Pei Wei
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| | - Tao Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
31
|
Graner MW, Lillehei KO, Katsanis E. Endoplasmic reticulum chaperones and their roles in the immunogenicity of cancer vaccines. Front Oncol 2015; 4:379. [PMID: 25610811 PMCID: PMC4285071 DOI: 10.3389/fonc.2014.00379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022] Open
Abstract
The endoplasmic reticulum (ER) is a major site of passage for proteins en route to other organelles, to the cell surface, and to the extracellular space. It is also the transport route for peptides generated in the cytosol by the proteasome into the ER for loading onto major histocompatibility complex class I (MHC I) molecules for eventual antigen presentation at the cell surface. Chaperones within the ER are critical for many of these processes; however, outside the ER certain of those chaperones may play important and direct roles in immune responses. In some cases, particular ER chaperones have been utilized as vaccines against tumors or infectious disease pathogens when purified from tumor tissue or recombinantly generated and loaded with antigen. In other cases, the cell surface location of ER chaperones has implications for immune responses as well as possible tumor resistance. We have produced heat-shock protein/chaperone protein-based cancer vaccines called “chaperone-rich cell lysate” (CRCL) that are conglomerates of chaperones enriched from solid tumors by an isoelectric focusing technique. These preparations have been effective against numerous murine tumors, as well as in a canine with an advanced lung carcinoma treated with autologous CRCL. We also published extensive proteomic analyses of CRCL prepared from human surgically resected tumor samples. Of note, these preparations contained at least 10 ER chaperones and a number of other residents, along with many other chaperones/heat-shock proteins. Gene ontology and network analyses utilizing these proteins essentially recapitulate the antigen presentation pathways and interconnections. In conjunction with our current knowledge of cell surface/extracellular ER chaperones, these data collectively suggest that a systems-level view may provide insight into the potent immune stimulatory activities of CRCL with an emphasis on the roles of ER components in those processes.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado School of Medicine , Aurora, CO , USA
| | - Kevin O Lillehei
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado School of Medicine , Aurora, CO , USA
| | - Emmanuel Katsanis
- Department of Pediatrics, The University of Arizona , Tucson, AZ , USA
| |
Collapse
|
32
|
Liu A, Ming JY, Fiskesund R, Ninio E, Karabina SA, Bergmark C, Frostegård AG, Frostegård J. Induction of dendritic cell-mediated T-cell activation by modified but not native low-density lipoprotein in humans and inhibition by annexin a5: involvement of heat shock proteins. Arterioscler Thromb Vasc Biol 2014; 35:197-205. [PMID: 25395618 DOI: 10.1161/atvbaha.114.304342] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Atherosclerosis is an inflammatory disease, where activated immunocompetent cells, including dendritic cells (DCs) and T cells are abundant in plaques. Low-density lipoprotein modified either by oxidation (oxLDL) or by human group X-secreted phospholipase A2 (LDLx) and heat shock proteins (HSP), especially HSP60 and 90, have been implicated in atherosclerosis. We previously reported that Annexin A5 inhibits inflammatory effects of phospholipids, decreases vascular inflammation and improves vascular function in apolipoprotein E(-/-) mice. Here, we focus on the LDLx effects on human DCs and T cells. APPROACH AND RESULTS Human DCs were differentiated from peripheral blood monocytes, stimulated by oxLDL or LDLx. Naive autologous T cells were cocultured with pretreated DCs. oxLDL and LDLx, in contrast to LDL, induced DC-activation and T-cell proliferation. T cells exposed to LDLx-treated DCs produced interferon-γ, interleukin (IL)-17 but not IL-4 and IL-10. Annexin A5 abrogated LDLx effects on DCs and T cells and increased production of transforming growth factor-β and IL-10. Furthermore, IL-10 producing T cells suppressed primary T-cell activation via soluble IL-10, transforming growth factor-β, and cell-cell contact. Lentiviral-mediated shRNA knock-down HSP60 and 90 in DCs attenuated the effect of LDLx on DCs and subsequent T-cell proliferation. Experiments on DC and T cells derived from carotid atherosclerotic plaques gave similar results. CONCLUSIONS Our data show that modified forms of LDL such as LDLx but not native LDL activate human T cells through DCs. HSP60 and 90 contribute to such T-cell activation. Annexin A5 promotes induction of regulatory T cells and is potentially interesting as a therapeutic agent.
Collapse
Affiliation(s)
- Anquan Liu
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.).
| | - Julia Yue Ming
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Roland Fiskesund
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Ewa Ninio
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Sonia-Athina Karabina
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Claes Bergmark
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Anna G Frostegård
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| | - Johan Frostegård
- From the Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.L., J.Y.M., R.F., A.G.F., J.F.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, ICAN, Genomics and Pathophysiology of Cardiovascular Diseases Team, Paris, France (E.N.); Sorbonne Universités, UPMC University Paris 06, INSERM UMR_933, Hôpital Armand-Trousseau, Paris, France (S.-A.K.); Division of Vascular Surgery, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (C.B.); and Division of Acute Internal Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden (J.F.)
| |
Collapse
|
33
|
Rapid isolation of extracellular vesicles from cell culture and biological fluids using a synthetic peptide with specific affinity for heat shock proteins. PLoS One 2014; 9:e110443. [PMID: 25329303 PMCID: PMC4201556 DOI: 10.1371/journal.pone.0110443] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/12/2014] [Indexed: 02/07/2023] Open
Abstract
Recent studies indicate that extracellular vesicles are an important source material for many clinical applications, including minimally-invasive disease diagnosis. However, challenges for rapid and simple extracellular vesicle collection have hindered their application. We have developed and validated a novel class of peptides (which we named venceremin, or Vn) that exhibit nucleotide-independent specific affinity for canonical heat shock proteins. The Vn peptides were validated to specifically and efficiently capture HSP-containing extracellular vesicles from cell culture growth media, plasma, and urine by electron microscopy, atomic force microscopy, sequencing of nucleic acid cargo, proteomic profiling, immunoblotting, and nanoparticle tracking analysis. All of these analyses confirmed the material captured by the Vn peptides was comparable to those purified by the standard ultracentrifugation method. We show that the Vn peptides are a useful tool for the rapid isolation of extracellular vesicles using standard laboratory equipment. Moreover, the Vn peptides are adaptable to diverse platforms and therefore represent an excellent solution to the challenge of extracellular vesicle isolation for research and clinical applications.
Collapse
|
34
|
Xu W, Xilinbaoleri, Liu H, Wang R, Bai J. Spinal cord biological safety of image-guided radiation therapy versus conventional radiation therapy. Neural Regen Res 2014; 7:2755-60. [PMID: 25317124 PMCID: PMC4190856 DOI: 10.3969/j.issn.1673-5374.2012.35.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/10/2012] [Indexed: 11/18/2022] Open
Abstract
Tumor models were simulated in purebred Beagles at the T9-10 levels of the spinal cord and treated with spinal image-guided radiation therapy or conventional radiation therapy with 50 or 70 Gy total radiation. Three months after radiation, neuronal injury at the T9-10 levels was observed, including reversible injury induced by spinal image-guided radiation therapy and apoptosis induced by conventional radiation therapy. The number of apoptotic cells and expression of the proapoptotic protein Fas were significantly reduced, but expression of the anti-apoptotic protein heat shock protein 70 was significantly increased after image-guided radiation therapy compared with the conventional method of the same radiation dose. Moreover, the spinal cord cell apoptotic index positively correlated with the ratio of Fas/heat shock protein 70. These findings indicate that 3 months of radiation therapy can induce a late response in the spinal cord to radiation therapy; image-guided radiation therapy is safer and results in less neuronal injury compared with conventional radiation therapy.
Collapse
Affiliation(s)
- Wanlong Xu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China ; People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Xilinbaoleri
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Hao Liu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Ruozheng Wang
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Jingping Bai
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
35
|
Pitt JM, Charrier M, Viaud S, André F, Besse B, Chaput N, Zitvogel L. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. THE JOURNAL OF IMMUNOLOGY 2014; 193:1006-11. [PMID: 25049431 DOI: 10.4049/jimmunol.1400703] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exosomes are nanometric membrane vesicles of late endosomal origin released by most, if not all, cell types as a means of sophisticated intercellular communication. A multitude of studies showed how exosomes can mediate and regulate immune responses against tumors. Dendritic cell-derived exosomes (Dex) have received much attention as immunotherapeutic anticancer agents since the discovery that they harbor functional MHC-peptide complexes, in addition to various other immune-stimulating components, that together facilitate immune cell-dependent tumor rejection. The therapeutic potential of Dex has been substantiated with their development and clinical testing in the treatment of cancer. This review focuses on mechanisms by which Dex interact with and influence immune cells and describes how they can be engineered to promote their immunogenic capacity as novel and dynamic anticancer agents.
Collapse
Affiliation(s)
- Jonathan M Pitt
- INSERM Unit U1015, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France
| | - Mélinda Charrier
- INSERM Unit U1015, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France
| | - Sophie Viaud
- INSERM Unit U1015, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France
| | - Fabrice André
- Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France; INSERM Unit U981, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Department of Medical Oncology, Gustave Roussy Cancer Campus, 98405 Villejuif, France
| | - Benjamin Besse
- Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France; Department of Cancer Medicine, Gustave Roussy Cancer Campus, 98405 Villejuif, France; and
| | - Nathalie Chaput
- INSERM Unit U1015, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France
| | - Laurence Zitvogel
- INSERM Unit U1015, Gustave Roussy Cancer Campus, 98405 Villejuif, France; Faculté de Médecine, Université Paris Sud-XI, 94276 Le Kremlin Bicêtre, France; Center of Clinical Investigations in Biotherapies of Cancer 507, Gustave Roussy Cancer Campus, 98405 Villejuif, France
| |
Collapse
|
36
|
Kakeda M, Arock M, Schlapbach C, Yawalkar N. Increased expression of heat shock protein 90 in keratinocytes and mast cells in patients with psoriasis. J Am Acad Dermatol 2014; 70:683-690.e1. [DOI: 10.1016/j.jaad.2013.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/28/2013] [Accepted: 12/02/2013] [Indexed: 11/25/2022]
|
37
|
Wolf JH, Bhatti TR, Fouraschen S, Chakravorty S, Wang L, Kurian S, Salomon D, Olthoff KM, Hancock WW, Levine MH. Heat shock protein 70 is required for optimal liver regeneration after partial hepatectomy in mice. Liver Transpl 2014; 20:376-85. [PMID: 24357103 PMCID: PMC3947447 DOI: 10.1002/lt.23813] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 11/21/2013] [Indexed: 01/05/2023]
Abstract
Liver regeneration is a complex process that restores functional tissue after resection or injury, and it is accompanied by transient adenosine triphosphate depletion and metabolic stress in hepatic parenchymal cells. Heat shock protein 70 (Hsp70) functions as a chaperone during periods of cellular stress and induces the expression of several inflammatory cytokines identified as key players during early liver regeneration. We, therefore, hypothesized that Hsp70 is required for the initiation of regeneration. Investigations were carried out in a 70% partial hepatectomy mouse model with mice lacking inducible Hsp70 (Hsp70(-/-)). Liver regeneration was assessed postoperatively with the liver weight/body weight (LW/BW) ratio, and sera and tissues were collected for analysis. In addition, the expression of Hsp-related genes was assessed in a cohort of 23 human living donor liver transplantation donors. In mice, the absence of Hsp70 was associated with a reduced postoperative LW/BW ratio, Ki-67 staining, and tumor necrosis factor α (TNF-α) expression in comparison with wild-type mice. TNF-α expression was also reduced in livers from Hsp70(-/-) mice after induction with lipopolysaccharide (1 mg/kg). Clinically, the transcription of multiple Hsp genes (especially Hsp70 family members) was up-regulated after donor hepatectomy. Together, these results suggest that the early phase of successful liver regeneration requires the presence of Hsp70 to induce TNF-α. Further studies are required to determine whether Hsp70 contributes to liver regeneration as a chaperone by stabilizing specific interactions required for growth signaling or as a paracrine inflammatory signal, as can occur in models of shock.
Collapse
Affiliation(s)
- Joshua H. Wolf
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Tricia R. Bhatti
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia/University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Suomi Fouraschen
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Shourjo Chakravorty
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia/University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | | | - Kim M. Olthoff
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia/University of Pennsylvania School of Medicine, Philadelphia, PA,Correspondence and proofs: Wayne W. Hancock, Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, 3615 Civic Ctr. Blvd., Philadelphia PA 19104, Telephone: (215) 590-8709, Fax: (215) 590-7384,
| | - Matthew H. Levine
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
38
|
Wang L, Yu Y. Dendritic cells primed with protein-protein fusion adjuvant. Methods Mol Biol 2014; 1139:57-75. [PMID: 24619671 DOI: 10.1007/978-1-4939-0345-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
To develop efficient T cell priming cancer vaccines, various recombinant fusion proteins have been developed by fusing a tumor antigen with a protein capable of stimulating or targeting dendritic cells (DC), the most important antigen-presenting cells for inducing CD8(+) cytotoxic T lymphocytes (CTL) which can efficiently kill tumor cells expressing the tumor antigen. The DC-stimulating or DC-targeting proteins, including granulocyte/macrophage colony-stimulating factor (GM-CSF), anti-DEC-205 monoclonal antibodies, flagellin, and heat shock proteins (HSP), function as promising intermolecular adjuvants. Herein, we describe in vitro assays on human DC pulsed with HSP fusion proteins, which might be useful in preclinical studies for the screening and assessment of candidate cancer vaccines.
Collapse
Affiliation(s)
- Liying Wang
- Norman Bethune College of Medicine, Jilin University, Changchun, China
| | | |
Collapse
|
39
|
Heil M, Land WG. Danger signals - damaged-self recognition across the tree of life. FRONTIERS IN PLANT SCIENCE 2014; 5:578. [PMID: 25400647 PMCID: PMC4215617 DOI: 10.3389/fpls.2014.00578] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 10/07/2014] [Indexed: 05/15/2023]
Abstract
Multicellular organisms suffer injury and serve as hosts for microorganisms. Therefore, they require mechanisms to detect injury and to distinguish the self from the non-self and the harmless non-self (microbial mutualists and commensals) from the detrimental non-self (pathogens). Danger signals are "damage-associated molecular patterns" (DAMPs) that are released from the disrupted host tissue or exposed on stressed cells. Seemingly ubiquitous DAMPs are extracellular ATP or extracellular DNA, fragmented cell walls or extracellular matrices, and many other types of delocalized molecules and fragments of macromolecules that are released when pre-existing precursors come into contact with enzymes from which they are separated in the intact cell. Any kind of these DAMPs enable damaged-self recognition, inform the host on tissue disruption, initiate processes aimed at restoring homeostasis, such as sealing the wound, and prepare the adjacent tissues for the perception of invaders. In mammals, antigen-processing and -presenting cells such as dendritic cells mature to immunostimulatory cells after the perception of DAMPs, prime naïve T-cells and elicit a specific adaptive T-/B-cell immune response. We discuss molecules that serve as DAMPs in multiple organisms and their perception by pattern recognition receptors (PRRs). Ca(2+)-fluxes, membrane depolarization, the liberation of reactive oxygen species and mitogen-activated protein kinase (MAPK) signaling cascades are the ubiquitous molecular mechanisms that act downstream of the PRRs in organisms across the tree of life. Damaged-self recognition contains both homologous and analogous elements and is likely to have evolved in all eukaryotic kingdoms, because all organisms found the same solutions for the same problem: damage must be recognized without depending on enemy-derived molecules and responses to the non-self must be directed specifically against detrimental invaders.
Collapse
Affiliation(s)
- Martin Heil
- Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-IrapuatoIrapuato, México
- *Correspondence: Martin Heil, Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Irapuato, Km 9.6 Libramiento Norte, Carretera Irapuato- León, Irapuato, Guanajuato, Mexico e-mail:
| | - Walter G. Land
- Molecular ImmunoRheumatology, INSERM UMR S1109, Laboratory of Excellence Transplantex, Faculty of Medicine, University of StrasbourgStrasbourg, France
| |
Collapse
|
40
|
Vanden Bergh P, Heller M, Braga-Lagache S, Frey J. The Aeromonas salmonicida subsp. salmonicida exoproteome: global analysis, moonlighting proteins and putative antigens for vaccination against furunculosis. Proteome Sci 2013; 11:44. [PMID: 24127837 PMCID: PMC3826670 DOI: 10.1186/1477-5956-11-44] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 10/04/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Aeromonas salmonicida subsp. salmonicida, the etiologic agent of furunculosis, is a major pathogen of fisheries worldwide. Despite the identification of several virulence factors the pathogenesis is still poorly understood. We have used high-throughput proteomics to display the differences between in vitro secretome of A. salmonicida wild-type (wt, hypervirulent, JF5054) and T3SS-deficient (isogenic ΔascV, extremely low-virulent, JF2747) strains in exponential (GP) and stationary (SP) phases of growth. RESULTS Among the different experimental conditions we obtained semi-quantitative values for a total of 2136 A. salmonicida proteins. Proteins of specific A. salmonicida species were proportionally less detected than proteins common to the Aeromonas genus or those shared with other Aeromonas species, suggesting that in vitro growth did not induce the expression of these genes. Four detected proteins which are unidentified in the genome of reference strains of A. salmonicida were homologous to components of the conjugative T4SS of A. hydrophila pRA1 plasmid. Polypeptides of three proteins which are specific to the 01-B526 strain were also discovered. In supernatants (SNs), the number of detected proteins was higher in SP (326 for wt vs 329 for mutant) than in GP (275 for wt vs 263 for mutant). In pellets, the number of identified proteins (a total of 1536) was approximately the same between GP and SP. Numerous highly conserved cytoplasmic proteins were present in A. salmonicida SNs (mainly EF-Tu, EF-G, EF-P, EF-Ts, TypA, AlaS, ribosomal proteins, HtpG, DnaK, peptidyl-prolyl cis-trans isomerases, GAPDH, Enolase, FbaA, TpiA, Pgk, TktA, AckA, AcnB, Mdh, AhpC, Tpx, SodB and PNPase), and several evidences support the theory that their extracellular localization was not the result of cell lysis. According to the Cluster of Orthologous Groups classification, 29% of excreted proteins in A. salmonicida SNs were currently poorly characterized. CONCLUSIONS In this part of our work we elucidated the whole in vitro exoproteome of hypervirulent A. salmonicida subsp. salmonicida and showed the secretion of several highly conserved cytoplasmic proteins with putative moonlighting functions and roles in virulence. All together, our results offer new information about the pathogenesis of furunculosis and point out potential candidates for vaccine development.
Collapse
Affiliation(s)
- Philippe Vanden Bergh
- Institute of Veterinary Bacteriology, University of Bern, Länggassstrasse 122, P.O. Box 8466, 3001 Bern, Switzerland
| | - Manfred Heller
- Department of Clinical Research, University of Bern, P.O. Box 37, 3010 Bern, Switzerland
| | - Sophie Braga-Lagache
- Department of Clinical Research, University of Bern, P.O. Box 37, 3010 Bern, Switzerland
| | - Joachim Frey
- Institute of Veterinary Bacteriology, University of Bern, Länggassstrasse 122, P.O. Box 8466, 3001 Bern, Switzerland
| |
Collapse
|
41
|
Du G, Liu Y, Li J, Liu W, Wang Y, Li H. Hypothermic microenvironment plays a key role in tumor immune subversion. Int Immunopharmacol 2013; 17:245-53. [DOI: 10.1016/j.intimp.2013.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/08/2013] [Accepted: 06/14/2013] [Indexed: 12/31/2022]
|
42
|
The Aeromonas salmonicida subsp. salmonicida exoproteome: determination of the complete repertoire of Type-Three Secretion System effectors and identification of other virulence factors. Proteome Sci 2013; 11:42. [PMID: 24073886 PMCID: PMC3852671 DOI: 10.1186/1477-5956-11-42] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/23/2013] [Indexed: 01/24/2023] Open
Abstract
Background Aeromonas salmonicida subsp. salmonicida, the etiologic agent of furunculosis, is a major pathogen of fisheries worldwide. Several virulence factors have been described, but the type-three secretion system (T3SS) is recognized as having a major effect on virulence by injecting effectors directly into fish cells. In this study we used high-throughput proteomics to display the differences between in vitro secretome of A. salmonicida wild-type (wt, hypervirulent, JF2267) and T3SS-deficient (isogenic ΔascV, extremely low-virulent, JF2747) strains in exponential and stationary phases of growth. Results Results confirmed the secretion of effectors AopH, AexT, AopP and AopO via T3SS, and for the first time demonstrated the impact of T3SS in secretion of Ati2, AopN and ExsE that are known as effectors in other pathogens. Translocators, needle subunits, Ati1, and AscX were also secreted in supernatants (SNs) dependent on T3SS. AopH, Ati2, AexT, AopB and AopD were in the top seven most abundant excreted proteins. EF-G, EF-Tu, DnaK, HtpG, PNPase, PepN and MdeA were moderately secreted in wt SNs and predicted to be putative T3 effectors by bioinformatics. Pta and ASA_P5G088 were increased in wt SNs and T3-associated in other bacteria. Ten conserved cytoplasmic proteins were more abundant in wt SNs than in the ΔascV mutant, but without any clear association to a secretion system. T1-secreted proteins were predominantly found in wt SNs: OmpAI, OmpK40, DegQ, insulinase ASA_0716, hypothetical ASA_0852 and ASA_3619. Presence of T3SS components in pellets was clearly decreased by ascV deletion, while no impact was observed on T1- and T2SS. Our results demonstrated that the ΔascV mutant strain excreted well-described (VapA, AerA, AerB, GCAT, Pla1, PlaC, TagA, Ahe2, GbpA and enolase) and yet uncharacterized potential toxins, adhesins and enzymes as much as or even more than the wt strain. Other putative important virulence factors were not detected. Conclusions We demonstrated the whole in vitro secretome and T3SS repertoire of hypervirulent A. salmonicida. Several toxins, adhesins and enzymes that are not part of the T3SS secretome were secreted to a higher extent in the extremely low-virulent ΔascV mutant. All together, our results show the high importance of an intact T3SS to initiate the furunculosis and offer new information about the pathogenesis.
Collapse
|
43
|
Dubaniewicz A. Microbial and human heat shock proteins as 'danger signals' in sarcoidosis. Hum Immunol 2013; 74:1550-8. [PMID: 23993988 DOI: 10.1016/j.humimm.2013.08.275] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/02/2013] [Accepted: 08/10/2013] [Indexed: 10/26/2022]
Abstract
In the light of the Matzinger's model of immune response, human heat shock proteins (HSPs) as main 'danger signals' (tissue damage-associated molecular patterns-DAMPs) or/and microbial HSPs as pathogen-associated molecular patterns (PAMPs) recognized by pattern recognition receptors (PRR), may induce sarcoid granuloma by both infectious and non-infectious factors in genetically different predisposed host. Regarding infectious causes of sarcoid models, low-virulence strains of, e.g. mycobacteria and propionibacteria recognized through changed PRR and persisting in altered host phagocytes, generate increased release of both human and microbial HSPs with their molecular and functional homology. High chronic spread of human and microbial HSPs altering cytokines, co-stimulatory molecules, and Tregs expression, apoptosis, oxidative stress, induces the autoimmunity, considered in sarcoidosis. Regarding non-infectious causes of sarcoidosis, human HSPs may be released at high levels during chronic low-grade exposure to misfolding amyloid precursor protein in stressed cells, phagocyted metal fumes, pigments with/without aluminum in tattoos, and due to heat shock in firefighters. Therefore, human HSPs as DAMPs and/or microbial HSPs as PAMPs produced as a result of non-infectious and infectious factors may induce different models of sarcoidosis, depending on the genetic background of the host. The number/expression of PRRs/ligands may influence the occurrence of sarcoidosis in particular organs.
Collapse
Affiliation(s)
- Anna Dubaniewicz
- Department of Pneumology, Medical University of Gdansk, Debinki 7 St., 80-211 Gdansk, Poland.
| |
Collapse
|
44
|
Graner MW, Romanoski A, Katsanis E. The 'peptidome' of tumour-derived chaperone-rich cell lysate anti-cancer vaccines reveals potential tumour antigens that stimulate tumour immunity. Int J Hyperthermia 2013; 29:380-9. [PMID: 23725202 DOI: 10.3109/02656736.2013.793406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tumour-derived chaperone-rich cell lysate (CRCL) when isolated from tumour tissue or when embedded with peptide antigens is a potent anti-cancer vaccine consisting of numerous chaperone/heat shock proteins, including the highly immunogenic Hsp70, Hsp90, glucose regulated protein 94, and calreticulin. We have previously documented that CRCL provides both a source of tumour antigens and danger signals triggering antigen presenting cell activation. In this report we describe the 'peptidome' of potential antigens extracted from CRCL prepared from a murine tumour. Using mass spectrometry techniques we identify almost 60 different proteins of origin for the CRCL peptides; we determine that the parental proteins come from essentially all parts of the cell, and are involved in a broad range of functions. Further in silico analysis demonstrates that the parental proteins are components of major signalling networks of vital importance for cancer cell survival, proliferation, and migration. In many instances the peptides identified possess amino acid sequences that would allow their putative binding and display by murine major histocompatibility complex class I and II molecules, and there are also predicted binding motifs for Hsp70-type chaperones. By mixing fractionated pools of peptides with antigen-free (normal liver) CRCL, we were able to reconstitute effective anti-tumour activity of the vaccine, showing that the peptides are indeed the major purveyors of CRCL vaccines' efficacy.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Neurosurgery, University of Colorado School of Medicine, Anschutz Medical Campus, Denver, CO 80045, USA.
| | | | | |
Collapse
|
45
|
Abstract
Atherosclerosis, the major cause of cardiovascular disease (CVD), is a chronic inflammatory condition with immune competent cells in lesions producing mainly pro-inflammatory cytokines. Dead cells and oxidized forms of low density lipoproteins (oxLDL) are abundant. The major direct cause of CVD appears to be rupture of atherosclerotic plaques. oxLDL has proinflammatory and immune-stimulatory properties, causes cell death at higher concentrations and contains inflammatory phospholipids with phosphorylcholine (PC) as an interesting epitope. Antibodies against PC (anti-PC) may be atheroprotective, one mechanism being anti-inflammatory. Bacteria and virus have been discussed, but it has been difficult to find direct evidence, and antibiotic trials have not been successful. Heat shock proteins could be one major target for atherogenic immune reactions. More direct causes of plaque rupture include pro-inflammatory cytokines, chemokines, and lipid mediators. To prove that inflammation is a cause of atherosclerosis and CVD, clinical studies with anti-inflammatory and/or immune-modulatory treatment are needed. The potential causes of immune reactions and inflammation in atherosclerosis and how inflammation can be targeted therapeutically to provide novel treatments for CVD are reviewed.
Collapse
Affiliation(s)
- Johan Frostegård
- Institute of Environmental Medicine, Unit of Immunology and Chronic Disease, Nobels väg 13, Stockholm, Sweden.
| |
Collapse
|
46
|
Abstract
Idiosyncratic drug reactions are a significant cause of morbidity and mortality for patients; they also markedly increase the uncertainty of drug development. The major targets are skin, liver, and bone marrow. Clinical characteristics suggest that IDRs are immune mediated, and there is substantive evidence that most, but not all, IDRs are caused by chemically reactive species. However, rigorous mechanistic studies are very difficult to perform, especially in the absence of valid animal models. Models to explain how drugs or reactive metabolites interact with the MHC/T-cell receptor complex include the hapten and P-I models, and most recently it was found that abacavir can interact reversibly with MHC to alter the endogenous peptides that are presented to T cells. The discovery of HLA molecules as important risk factors for some IDRs has also significantly contributed to our understanding of these adverse reactions, but it is not yet clear what fraction of IDRs have a strong HLA dependence. In addition, with the exception of abacavir, most patients who have the HLA that confers a higher IDR risk with a specific drug will not have an IDR when treated with that drug. Interindividual differences in T-cell receptors and other factors also presumably play a role in determining which patients will have an IDR. The immune response represents a delicate balance, and immune tolerance may be the dominant response to a drug that can cause IDRs.
Collapse
Affiliation(s)
- Jack Uetrecht
- Faculties of Pharmacy and Medicine, University of Toronto, Toronto, Canada M5S3M2.
| | | |
Collapse
|