1
|
Wang HH, He LL, Zhang J, Hua BZ. Sexual dimorphism and ultrastructure of salivary glands in adult Bittacus cirratus Tjeder, 1956 (Mecoptera: Bittacidae). ARTHROPOD STRUCTURE & DEVELOPMENT 2025; 86:101441. [PMID: 40220630 DOI: 10.1016/j.asd.2025.101441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/14/2025]
Abstract
Sexual dimorphism is ubiquitous in insects and has been extensively studied across various taxa. However, it remains unclear whether the salivary glands exhibit sexual dimorphism in adult Bittacidae. In this study, a comparative study of salivary glands in adult male and female Bittacus cirratus Tjeder, 1956 was performed using light and transmission electron microscopy. The salivary glands are tubular labial glands in both sexes of B. cirratus, consisting of a salivary pump, a common salivary duct, a pair of efferent ducts, and numerous dichotomously branching secretory tubules. Interestingly, the salivary glands in females contain a pair of small salivary reservoirs, which are not present in the males. The male salivary glands have a significantly larger diameter and more secretory tubules than those in females. In contrast, male secretory cells occupy a minimal volume of the secretory tubule, resulting in a larger secretory lumen that nearly fills the entire tubule. This configuration enhances the storage capacity of the male secretory tubules. Male secretory cells exhibit active autophagy, possibly due to their approaching late life stages. Sexual dimorphism in the salivary glands and the possible reasons are briefly discussed in Bittacidae.
Collapse
Affiliation(s)
- He-Hong Wang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Le-Le He
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jie Zhang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Bao-Zhen Hua
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
2
|
Luo R, Wang Z, Xu F, Xie K. Dexmedetomidine improve lung inflammation by regulating autophagy and apoptosis of CD4+ T cell via AMPK/mTOR signaling. Mol Immunol 2025; 183:1-11. [PMID: 40311186 DOI: 10.1016/j.molimm.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES To investigate the protective effect and potential mechanism of dexmedetomidine (Dex) in acute lung injury (ALI). MATERIALS AND METHODS C57BL/6 mice and EL-4 cells were used for in vivo and in vitro studies, respectively. Cecal ligation and puncture (CLP) method was used to prepare an acute lung injury model. After dexmedetomidine intervention, tissue and cell samples were collected to evaluate and measure the severity of lung damage, the proportion of Treg cells, the expression of autophagy-related protein levels and AMPK/mTOR pathways. RESULTS Dex reduced lung damage, and IL-17a, MPO positive cells in the lung, decreased the levels of pro-inflammatory cytokines, and restrain autophagy and apoptosis via the activation of the AMPK/mTOR pathway and increase of the proportion of Tregs. CONCLUSIONS Dex can inhibit the levels of autophagy and apoptosis, increase the proportion of Treg cells, and reduce CLP induced acute lung injury through regulating AKMP/MTOR pathway.
Collapse
Affiliation(s)
- Renjie Luo
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Zhao Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Ke Xie
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Shabnum SS, Siranjeevi R, Raj CK, Saravanan A, Vickram AS, Chopra H, Malik T. Advancements in nanotechnology-driven photodynamic and photothermal therapies: mechanistic insights and synergistic approaches for cancer treatment. RSC Adv 2024; 14:38952-38995. [PMID: 39659608 PMCID: PMC11629304 DOI: 10.1039/d4ra07114j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer is a disease that involves uncontrolled cell division triggered by genetic damage to the genes that control cell growth and division. Cancer starts as a localized illness, but subsequently spreads to other areas in the human body (metastasis), making it incurable. Cancer is the second most prevalent cause of mortality worldwide. Every year, almost ten million individuals get diagnosed with cancer. Although different cancer treatment options exist, such as chemotherapy, radiation, surgery and immunotherapy, their clinical efficacy is limited due to their significant side effects. New cancer treatment options, such as phototherapy, which employs light for the treatment of cancer, have sparked a growing fascination in the cancer research community. Phototherapies are classified into two types: photodynamic treatment (PDT) and photothermal therapy (PTT). PDT necessitates the use of a photosensitizing chemical and exposure to light at a certain wavelength. Photodynamic treatment (PDT) is primarily based on the creation of singlet oxygen by the stimulation of a photosensitizer, which is then used to kill tumor cells. PDT can be used to treat a variety of malignancies. On the other hand, PTT employs a photothermal molecule that activates and destroys cancer cells at the longer wavelengths of light, making it less energetic and hence less hazardous to other cells and tissues. While PTT is a better alternative to standard cancer therapy, in some irradiation circumstances, it can cause cellular necrosis, which results in pro-inflammatory reactions that can be harmful to therapeutic effectiveness. Latest research has revealed that PTT may be adjusted to produce apoptosis instead of necrosis, which is attractive since apoptosis reduces the inflammatory response.
Collapse
Affiliation(s)
- S Sameera Shabnum
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - R Siranjeevi
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - C Krishna Raj
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - A Saravanan
- Department of Sustainable Engineering, Institute of Biotechnology, Saveetha School of Engineering, SIMATS Chennai-602105 Tamil Nadu India
| | - A S Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University Rajpura 140401 Punjab India
| | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University 378 Jimma Ethiopia
- Division of Research & Development, Lovely Professional University Phagwara 144411 India
| |
Collapse
|
4
|
Hao M, Sebag SC, Qian Q, Yang L. Lysosomal physiology and pancreatic lysosomal stress in diabetes mellitus. EGASTROENTEROLOGY 2024; 2:e100096. [PMID: 39512752 PMCID: PMC11542681 DOI: 10.1136/egastro-2024-100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Endocrine and exocrine functions of the pancreas control nutritional absorption, utilisation and systemic metabolic homeostasis. Under basal conditions, the lysosome is pivotal in regulating intracellular organelles and metabolite turnover. In response to acute or chronic stress, the lysosome senses metabolic flux and inflammatory challenges, thereby initiating the adaptive programme to re-establish cellular homeostasis. A growing body of evidence has demonstrated the pathophysiological relevance of the lysosomal stress response in metabolic diseases in diverse sets of tissues/organs, such as the liver and the heart. In this review, we discuss the pathological relevance of pancreatic lysosome stress in diabetes mellitus. We begin by summarising lysosomal biology, followed by exploring the immune and metabolic functions of lysosomes and finally discussing the interplay between lysosomal stress and the pathogenesis of pancreatic diseases. Ultimately, our review aims to enhance our understanding of lysosomal stress in disease pathogenesis, which could potentially lead to the discovery of innovative treatment methods for these conditions.
Collapse
Affiliation(s)
- Meihua Hao
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Sara C Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
5
|
Yu Q, Ding J, Li S, Li Y. Autophagy in cancer immunotherapy: Perspective on immune evasion and cell death interactions. Cancer Lett 2024; 590:216856. [PMID: 38583651 DOI: 10.1016/j.canlet.2024.216856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Both the innate and adaptive immune systems work together to produce immunity. Cancer immunotherapy is a novel approach to tumor suppression that has arisen in response to the ineffectiveness of traditional treatments like radiation and chemotherapy. On the other hand, immune evasion can diminish immunotherapy's efficacy. There has been a lot of focus in recent years on autophagy and other underlying mechanisms that impact the possibility of cancer immunotherapy. The primary feature of autophagy is the synthesis of autophagosomes, which engulf cytoplasmic components and destroy them by lysosomal degradation. The planned cell death mechanism known as autophagy can have opposite effects on carcinogenesis, either increasing or decreasing it. It is autophagy's job to maintain the balance and proper functioning of immune cells like B cells, T cells, and others. In addition, autophagy controls whether macrophages adopt the immunomodulatory M1 or M2 phenotype. The ability of autophagy to control the innate and adaptive immune systems is noteworthy. Interleukins and chemokines are immunological checkpoint chemicals that autophagy regulates. Reducing antigen presentation to induce immunological tolerance is another mechanism by which autophagy promotes cancer survival. Therefore, targeting autophagy is of importance for enhancing potential of cancer immunotherapy.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jiajun Ding
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shisen Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yunlong Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Kasten-Jolly J, Lawrence DA. Cellular and Molecular Immunity to Influenza Viruses and Vaccines. Vaccines (Basel) 2024; 12:389. [PMID: 38675771 PMCID: PMC11154265 DOI: 10.3390/vaccines12040389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Immune responses to influenza (flu) antigens reflect memory of prior infections or vaccinations, which might influence immunity to new flu antigens. Memory of past antigens has been termed "original antigenic sin" or, more recently, "immune imprinting" and "seniority". We have researched a comparison between the immune response to live flu infections and inactivated flu vaccinations. A brief history of antibody generation theories is presented, culminating in new findings about the immune-network theory and suggesting that a network of clones exists between anti-idiotypic antibodies and T cell receptors. Findings regarding the 2009 pandemic flu strain and immune responses to it are presented, including memory B cells and conserved regions within the hemagglutinin protein. The importance of CD4+ memory T cells and cytotoxic CD8+ T cells responding to both infections and vaccinations are discussed and compared. Innate immune cells, like natural killer (NK) cells and macrophages, are discussed regarding their roles in adaptive immune responses. Antigen presentation via macroautophagy processes is described. New vaccines in development are mentioned along with the results of some clinical trials. The manuscript concludes with how repeated vaccinations are impacting the immune system and a sketch of what might be behind the imprinting phenomenon, including future research directions.
Collapse
Affiliation(s)
- Jane Kasten-Jolly
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA;
| | - David A. Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA;
- Departments of Biomedical Science and Environmental Health Science, University at Albany School of Public Health, Rensselaer, NY 12144, USA
| |
Collapse
|
7
|
Bhatia D, Choi ME. Autophagy and mitophagy: physiological implications in kidney inflammation and diseases. Am J Physiol Renal Physiol 2023; 325:F1-F21. [PMID: 37167272 PMCID: PMC10292977 DOI: 10.1152/ajprenal.00012.2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Autophagy is a ubiquitous intracellular cytoprotective quality control program that maintains cellular homeostasis by recycling superfluous cytoplasmic components (lipid droplets, protein, or glycogen aggregates) and invading pathogens. Mitophagy is a selective form of autophagy that by recycling damaged mitochondrial material, which can extracellularly act as damage-associated molecular patterns, prevents their release. Autophagy and mitophagy are indispensable for the maintenance of kidney homeostasis and exert crucial functions during both physiological and disease conditions. Impaired autophagy and mitophagy can negatively impact the pathophysiological state and promote its progression. Autophagy helps in maintaining structural integrity of the kidney. Mitophagy-mediated mitochondrial quality control is explicitly critical for regulating cellular homeostasis in the kidney. Both autophagy and mitophagy attenuate inflammatory responses in the kidney. An accumulating body of evidence highlights that persistent kidney injury-induced oxidative stress can contribute to dysregulated autophagic and mitophagic responses and cell death. Autophagy and mitophagy also communicate with programmed cell death pathways (apoptosis and necroptosis) and play important roles in cell survival by preventing nutrient deprivation and regulating oxidative stress. Autophagy and mitophagy are activated in the kidney after acute injury. However, their aberrant hyperactivation can be deleterious and cause tissue damage. The findings on the functions of autophagy and mitophagy in various models of chronic kidney disease are heterogeneous and cell type- and context-specific dependent. In this review, we discuss the roles of autophagy and mitophagy in the kidney in regulating inflammatory responses and during various pathological manifestations.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
8
|
Pujhari S, Heu CC, Brustolin M, Johnson RM, Kim D, Rasgon JL. Sindbis virus is suppressed in the yellow fever mosquito Aedes aegypti by ATG-6/Beclin-1 mediated activation of autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526867. [PMID: 36778292 PMCID: PMC9915692 DOI: 10.1101/2023.02.02.526867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a critical modulator of pathogen invasion response in vertebrates and invertebrates. However, how it affects mosquito-borne viral pathogens that significantly burden public health remains underexplored. To address this gap, we use a genetic approach to activate macroautophagy/autophagy in the yellow fever mosquito (Aedes aegypti), infected with a recombinant Sindbis virus (SINV) expressing an autophagy activator. We first demonstrate a 17-amino acid peptide derived from the Ae. aegypti autophagy-related protein 6 (ATG-6/beclin-1-like protein) is sufficient to induce autophagy in C6/36 mosquito cells, as marked by lipidation of ATG-8 and puncta formation. Next, we engineered a recombinant SINV expressing this bioactive beclin-1-like peptide and used it to infect and induce autophagy in adult mosquitoes. We find that modulation of autophagy using this recombinant SINV negatively regulated production of infectious viruses. The results from this study improve our understanding of the role of autophagy in arboviruses in invertebrate hosts and also highlight the potential for the autophagy pathway to be exploited for arboviral control.
Collapse
Affiliation(s)
- Sujit Pujhari
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina, Columbia, SC
| | - Chan C. Heu
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- USDA-ARS, Maricopa, AZ, USA
| | - Marco Brustolin
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- Unit of Entomology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Rebecca M. Johnson
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Entomology, The Connecticut Agricultural Experiment Station, New Haven, CT
| | - Donghun Kim
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Vector Entomology, Kyungpook National University, Daegu, South Korea
| | - Jason L. Rasgon
- Department of Entomology, the Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
9
|
Fonderflick L, Baudu T, Adotévi O, Guittaut M, Adami P, Delage-Mourroux R. The ATG8 Family Proteins GABARAP and GABARAPL1 Target Antigen to Dendritic Cells to Prime CD4+ and CD8+ T Cells. Cells 2022; 11:cells11182782. [PMID: 36139357 PMCID: PMC9496894 DOI: 10.3390/cells11182782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccine therapy is a promising method of research to promote T cell immune response and to develop novel antitumor immunotherapy protocols. Accumulating evidence has shown that autophagy is involved in antigen processing and presentation to T cells. In this work, we investigated the potential role of GABARAP and GABARAPL1, two members of the autophagic ATG8 family proteins, as surrogate tumor antigen delivery vectors to prime antitumor T cells. We showed that bone marrow-derived dendritic cells, expressing the antigen OVALBUMIN (OVA) fused with GABARAP or GABARAPL1, were able to prime OVA-specific CD4+ T cells in vitro. Interestingly, the fusion proteins were also degraded by the proteasome pathway and the resulting peptides were presented by the MHC class I system. We then asked if the aforementioned fusion proteins could improve tumor cell immunogenicity and T cell priming. The B16-F10 melanoma was chosen as the tumor cell line to express the fusion proteins. B16-F10 cells that expressed the OVA-ATG8 fused proteins stimulated OVA-specific CD8+ T cells, but demonstrated no CD4+ T cell response. In the future, these constructions may be used in vaccination trials as potential candidates to control tumor growth.
Collapse
Affiliation(s)
- Leïla Fonderflick
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
| | - Timothée Baudu
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
| | - Olivier Adotévi
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
- Departement of Oncology, Centre Hospitalier de Recherche Universitaire de Besançon, University Bourgogne Franche-Comté, 25000 Besançon, France
| | - Michaël Guittaut
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
- DImaCell Platform, University Bourgogne Franche-Comté, 25000 Besançon, France
| | - Pascale Adami
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
- Correspondence: (P.A.); (R.D.-M.)
| | - Régis Delage-Mourroux
- INSERM, EFS BFC, UMR1098, RIGHT Institute, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Group TIM-C, University Bourgogne Franche-Comté, 25000 Besançon, France
- Correspondence: (P.A.); (R.D.-M.)
| |
Collapse
|
10
|
Li J, Yu C, Shen F, Cui B, Liu N, Zhuang S. Class IIa histone deacetylase inhibition ameliorates acute kidney injury by suppressing renal tubular cell apoptosis and enhancing autophagy and proliferation. Front Pharmacol 2022; 13:946192. [PMID: 35935816 PMCID: PMC9354984 DOI: 10.3389/fphar.2022.946192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
Expression and function of histone deacetylases (HDACs) vary with cell types and pathological conditions. Our recent studies showed that pharmacological targeting class IIa HDACs attenuated renal fibrosis, but the effect of class IIa HDAC inhibition on acute kidney injury (AKI) remains unknown. In this study, we found that four class IIa HDACs (4, 5, 7, 9) were highly expressed in the kidney of folic acid (FA) and ischemia/reperfusion (I/R)-induced AKI in mice. Administration of TMP269, a potent and selective class IIa HDAC inhibitor, improved renal function and reduced tubular cell injury and apoptosis, with concomitant suppression of HDAC4 and elevation of acetyl-histone H3. Mechanistical studies showed that TMP269 treatment inhibited FA and I/R-induced caspase-3 cleavage, Bax expression and p53 phosphorylation. Conversely, TMP269 administration preserved expression of E-cadherin, BMP7, Klotho and Bcl-2 in injured kidneys. Moreover, TMP269 was effective in promoting cellular autophagy as indicated by increased expression of Atg7, beclin-1, and LC3II, and promoted renal tubular cell proliferation as shown by increased number of proliferating cell nuclear antigen-positive cells and expression of cyclin E. Finally, blocking class IIa HDACs inhibited FA-and I/R-induced phosphorylation of extracellular signal-regulated kinases 1 and 2, and p38, two signaling pathways associated with the pathogenesis of AKI. Collectively, these results suggest that pharmacological inhibition of class IIa HDACs protects against AKI through ameliorating apoptosis, enhancing autophagy and promoting proliferation of renal tubular cells by targeting multiple signaling pathways.
Collapse
Affiliation(s)
- Jialu Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Yu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fengchen Shen
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Binbin Cui
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Kawasaki T, Ikegawa M, Kawai T. Antigen Presentation in the Lung. Front Immunol 2022; 13:860915. [PMID: 35615351 PMCID: PMC9124800 DOI: 10.3389/fimmu.2022.860915] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
The lungs are constantly exposed to environmental and infectious agents such as dust, viruses, fungi, and bacteria that invade the lungs upon breathing. The lungs are equipped with an immune defense mechanism that involves a wide variety of immunological cells to eliminate these agents. Various types of dendritic cells (DCs) and macrophages (MACs) function as professional antigen-presenting cells (APCs) that engulf pathogens through endocytosis or phagocytosis and degrade proteins derived from them into peptide fragments. During this process, DCs and MACs present the peptides on their major histocompatibility complex class I (MHC-I) or MHC-II protein complex to naïve CD8+ or CD4+ T cells, respectively. In addition to these cells, recent evidence supports that antigen-specific effector and memory T cells are activated by other lung cells such as endothelial cells, epithelial cells, and monocytes through antigen presentation. In this review, we summarize the molecular mechanisms of antigen presentation by APCs in the lungs and their contribution to immune response.
Collapse
Affiliation(s)
| | | | - Taro Kawai
- *Correspondence: Takumi Kawasaki, ; Taro Kawai,
| |
Collapse
|
12
|
Chawla K, Subramanian G, Rahman T, Fan S, Chakravarty S, Gujja S, Demchak H, Chakravarti R, Chattopadhyay S. Autophagy in Virus Infection: A Race between Host Immune Response and Viral Antagonism. IMMUNO 2022; 2:153-169. [PMID: 35252965 PMCID: PMC8893043 DOI: 10.3390/immuno2010012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Virus-infected cells trigger a robust innate immune response and facilitate virus replication. Here, we review the role of autophagy in virus infection, focusing on both pro-viral and anti-viral host responses using a select group of viruses. Autophagy is a cellular degradation pathway operated at the basal level to maintain homeostasis and is induced by external stimuli for specific functions. The degradative function of autophagy is considered a cellular anti-viral immune response. However, autophagy is a double-edged sword in viral infection; viruses often benefit from it, and the infected cells can also use it to inhibit viral replication. In addition to viral regulation, autophagy pathway proteins also function in autophagy-independent manners to regulate immune responses. Since viruses have co-evolved with hosts, they have developed ways to evade the anti-viral autophagic responses of the cells. Some of these mechanisms are also covered in our review. Lastly, we conclude with the thought that autophagy can be targeted for therapeutic interventions against viral diseases.
Collapse
Affiliation(s)
- Karan Chawla
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Gayatri Subramanian
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Tia Rahman
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Shumin Fan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sukanya Chakravarty
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Shreyas Gujja
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Hayley Demchak
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ritu Chakravarti
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Correspondence:
| |
Collapse
|
13
|
Bhatti SI, Mindikoglu AL. The impact of dawn to sunset fasting on immune system and its clinical significance in Covid-19 pandemic. Metabol Open 2021; 13:100162. [PMID: 34977523 PMCID: PMC8713419 DOI: 10.1016/j.metop.2021.100162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Dawn to sunset fasting, a type of intermittent fasting commonly practiced in the month of Ramadan, requires fasting from dawn to sunset without food or liquid intake. Dawn and dusk are two transition time zones of the day that play a critical role in the human circadian rhythm. Practicing dawn to sunset fasting requires the alignment of mealtimes and wake-sleep times with the human biological dawn and dusk. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) impairs immune cell responses at multiple levels and leads to severe Coronavirus Disease 2019 (COVID-19). It generates high levels of pro-inflammatory cytokines and chemokines, also known as a cytokine storm, leads to mitochondrial dysfunction and generation of excessive amounts of mitochondrial reactive oxygen species, downregulates autophagy to escape detection for unchecked replication, and alters gut microbiome composition. Severe cases of COVID-19 have been associated with several comorbidities that impair immune responses (e.g., obesity, diabetes, malignancy) and blood laboratory abnormalities (e.g., elevated procalcitonin, C-reactive protein, interleukin-6 (IL-6), leukocytosis, lymphopenia). Several studies of dawn to sunset fasting showed anti-inflammatory effect by suppressing several pro-inflammatory cytokines, reducing oxidative stress, inducing a proteome response associated with increased autophagy, remodeling the gut microbiome, and improving the components of metabolic syndrome (e.g., obesity, blood glucose levels, blood pressure, lipids). In conclusion, dawn to sunset fasting has the potential to optimize the immune system function against SARS-CoV-2 during the COVID-19 pandemic as it suppresses chronic inflammation and oxidative stress, improves metabolic profile, and remodels the gut microbiome. This review presents scientific literature related to the effects of dawn to sunset fasting on the immune system. Studies are needed to assess and confirm the potential benefits of dawn to sunset fasting against SARS-CoV-2.
Collapse
Affiliation(s)
- Sundus I Bhatti
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| | - Ayse L Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021; 11:603224. [PMID: 33763351 PMCID: PMC7982729 DOI: 10.3389/fonc.2021.603224] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
Affiliation(s)
- Alejandra Suares
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Victoria Medina
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Omar Coso
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021. [DOI: 10.3389/fonc.2021.603224
expr 816899697 + 824303767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
|
16
|
Lee E, Sandgren K, Duette G, Stylianou VV, Khanna R, Eden JS, Blyth E, Gottlieb D, Cunningham AL, Palmer S. Identification of SARS-CoV-2 Nucleocapsid and Spike T-Cell Epitopes for Assessing T-Cell Immunity. J Virol 2021; 95:e02002-20. [PMID: 33443088 PMCID: PMC8579755 DOI: 10.1128/jvi.02002-20] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
Developing optimal T-cell response assays to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is critical for measuring the duration of immunity to this disease and assessing the efficacy of vaccine candidates. These assays need to target conserved regions of SARS-CoV-2 global variants and avoid cross-reactivity to seasonal human coronaviruses. To contribute to this effort, we employed an in silico immunoinformatics analysis pipeline to identify immunogenic peptides resulting from conserved and highly networked regions with topological importance from the SARS-CoV-2 nucleocapsid and spike proteins. A total of 57 highly networked T-cell epitopes that are conserved across geographic viral variants were identified from these viral proteins, with a binding potential to diverse HLA alleles and 80 to 100% global population coverage. Importantly, 18 of these T-cell epitope derived peptides had limited homology to seasonal human coronaviruses making them promising candidates for SARS-CoV-2-specific T-cell immunity assays. Moreover, two of the NC-derived peptides elicited effector/polyfunctional responses of CD8+ T cells derived from SARS-CoV-2 convalescent patients.IMPORTANCE The development of specific and validated immunologic tools is critical for understanding the level and duration of the cellular response induced by SARS-CoV-2 infection and/or vaccines against this novel coronavirus disease. To contribute to this effort, we employed an immunoinformatics analysis pipeline to define 57 SARS-CoV-2 immunogenic peptides within topologically important regions of the nucleocapsid (NC) and spike (S) proteins that will be effective for detecting cellular immune responses in 80 to 100% of the global population. Our immunoinformatics analysis revealed that 18 of these peptides had limited homology to circulating seasonal human coronaviruses and therefore are promising candidates for distinguishing SARS-CoV-2-specific immune responses from pre-existing coronavirus immunity. Importantly, CD8+ T cells derived from SARS-CoV-2 survivors exhibited polyfunctional effector responses to two novel NC-derived peptides identified as HLA-binders. These studies provide a proof of concept that our immunoinformatics analysis pipeline identifies novel immunogens which can elicit polyfunctional SARS-CoV-2-specific T-cell responses.
Collapse
Affiliation(s)
- Eunok Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Kerrie Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Gabriel Duette
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Vicki V Stylianou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Rajiv Khanna
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John-Sebastian Eden
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Emily Blyth
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- BMT and Cell Therapies Program, Westmead Hospital, Westmead, New South Wales, Australia
| | - David Gottlieb
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- BMT and Cell Therapies Program, Westmead Hospital, Westmead, New South Wales, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
17
|
Gonçalves RDC, Freire PP, Coletti D, Seelaender M. Tumor Microenvironment Autophagic Processes and Cachexia: The Missing Link? Front Oncol 2021; 10:617109. [PMID: 33604297 PMCID: PMC7884816 DOI: 10.3389/fonc.2020.617109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022] Open
Abstract
Cachexia is a syndrome that affects the entire organism and presents a variable plethora of symptoms in patients, always associated with continuous and involuntary degradation of skeletal muscle mass and function loss. In cancer, this syndrome occurs in 50% of all patients, while prevalence increases to 80% as the disease worsens, reducing quality of life, treatment tolerance, therapeutic response, and survival. Both chronic systemic inflammation and immunosuppression, paradoxically, correspond to important features in cachexia patients. Systemic inflammation in cachexia is fueled by the interaction between tumor and peripheral tissues with significant involvement of infiltrating immune cells, both in the peripheral tissues and in the tumor itself. Autophagy, as a process of regulating cellular metabolism and homeostasis, can interfere with the metabolic profile in the tumor microenvironment. Under a scenario of balanced autophagy in the tumor microenvironment, the infiltrating immune cells control cytokine production and secretion. On the other hand, when autophagy is unbalanced or dysfunctional within the tumor microenvironment, there is an impairment in the regulation of immune cell’s inflammatory phenotype. The inflammatory phenotype upregulates metabolic consumption and cytokine production, not only in the tumor microenvironment but also in other tissues and organs of the host. We propose that cachexia-related chronic inflammation can be, at least, partly associated with the failure of autophagic processes in tumor cells. Autophagy endangers tumor cell viability by producing immunogenic tumor antigens, thus eliciting the immune response necessary to counteract tumor progression, while preventing the establishment of inflammation, a hallmark of cachexia. Comprehensive understanding of this complex functional dichotomy may enhance cancer treatment response and prevent/mitigate cancer cachexia. This review summarizes the recent available literature regarding the role of autophagy within the tumor microenvironment and the consequences eliciting the development of cancer cachexia.
Collapse
Affiliation(s)
- Renata de Castro Gonçalves
- Cancer Metabolism Research Group, Department of Surgery, LIM26-HC, Faculdade de Medicina, and Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dario Coletti
- Sorbonne Université, CNRS UMR 8256, Inserm U1164, Biological Adaptation and Aging (B2A), Paris, France.,Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Rome, Italy
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery, LIM26-HC, Faculdade de Medicina, and Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Islam MN, Hossain KS, Sarker PP, Ferdous J, Hannan MA, Rahman MM, Chu DT, Uddin MJ. Revisiting pharmacological potentials of Nigella sativa seed: A promising option for COVID-19 prevention and cure. Phytother Res 2020; 35:1329-1344. [PMID: 33047412 PMCID: PMC7675410 DOI: 10.1002/ptr.6895] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/08/2020] [Accepted: 09/17/2020] [Indexed: 01/08/2023]
Abstract
Nigella sativa seed and its active compounds have been historically recognized as an effective herbal panacea that can establish a balanced inflammatory response by suppressing chronic inflammation and promoting healthy immune response. The essential oil and other preparations of N. sativa seed have substantial therapeutic outcomes against immune disturbance, autophagy dysfunction, oxidative stress, ischemia, inflammation, in several COVID‐19 comorbidities such as diabetes, cardiovascular disorders, Kawasaki‐like diseases, and many bacterial and viral infections. Compelling evidence in the therapeutic efficiency of N. sativa along with the recent computational findings is strongly suggestive of combating emerged COVID‐19 pandemic. Also, being an available candidate in nutraceuticals, N. sativa seed oil could be immensely potential and feasible to prevent and cure COVID‐19. This review was aimed at revisiting the pharmacological benefits of N. sativa seed and its active metabolites that may constitute a potential basis for developing a novel preventive and therapeutic strategy against COVID‐19. Bioactive compounds of N. sativa seed, especially thymiquinone, α‐hederin, and nigellidine, could be alternative and promising herbal drugs to combat COVID‐19. Preclinical and clinical trials are required to delineate detailed mechanism of N. sativa's active components and to investigate their efficacy and potency under specific pathophysiological conditions of COVID‐19.
Collapse
Affiliation(s)
- Mohammad Nazrul Islam
- Department of Biotechnology, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh.,ABEx Bio-Research Center, Dhaka, Bangladesh
| | - Khandkar Shaharina Hossain
- ABEx Bio-Research Center, Dhaka, Bangladesh.,Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Partha Protim Sarker
- ABEx Bio-Research Center, Dhaka, Bangladesh.,Mawlana Bhashani Science and Technology University Santosh, Tangail, Bangladesh
| | - Jannatul Ferdous
- ABEx Bio-Research Center, Dhaka, Bangladesh.,Department of Physiology, Biochemistry and Pharmacology, Chottogram Veterinary and Animal Science University, Chottogram, Bangladesh
| | - Md Abdul Hannan
- ABEx Bio-Research Center, Dhaka, Bangladesh.,Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh.,Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Md Masudur Rahman
- Department of Pathology, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Dinh-Toi Chu
- Hanoi National University of Education, Hanoi, Vietnam
| | - Md Jamal Uddin
- ABEx Bio-Research Center, Dhaka, Bangladesh.,Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Hannan MA, Rahman MA, Rahman MS, Sohag AAM, Dash R, Hossain KS, Farjana M, Uddin MJ. Intermittent fasting, a possible priming tool for host defense against SARS-CoV-2 infection: Crosstalk among calorie restriction, autophagy and immune response. Immunol Lett 2020; 226:38-45. [PMID: 32659267 PMCID: PMC7351063 DOI: 10.1016/j.imlet.2020.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/23/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative pathogen of deadly Coronavirus disease-19 (COVID-19) pandemic, which emerged as a major threat to public health across the world. Although there is no clear gender or socioeconomic discrimination in the incidence of COVID-19, individuals who are older adults and/or with comorbidities and compromised immunity have a relatively higher risk of contracting this disease. Since no specific drug has yet been discovered, strengthening immunity along with maintaining a healthy living is the best way to survive this disease. As a healthy practice, calorie restriction in the form of intermittent fasting (IF) in several clinical settings has been reported to promote several health benefits, including priming of the immune response. This dietary restriction also activates autophagy, a cell surveillance system that boosts up immunity. With these prevailing significance in priming host defense, IF could be a potential strategy amid this outbreak to fighting off SARS-CoV-2 infection. Currently, no review so far available proposing IF as an encouraging strategy in the prevention of COVID-19. A comprehensive review has therefore been planned to highlight the beneficial role of fasting in immunity and autophagy, that underlie the possible defense against SARS-CoV-2 infection. The COVID-19 pathogenesis and its impact on host immune response have also been briefly outlined. This review aimed at revisiting the immunomodulatory potential of IF that may constitute a promising preventive approach against COVID-19.
Collapse
Affiliation(s)
- Md. Abdul Hannan
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh,ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh,Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Md. Ataur Rahman
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh,Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea,Global Biotechnology & Biomedical Research Network (GBBRN), Dept. of Biotechnology and Genetic Engineering, Islamic University, Kushtia, 7003, Bangladesh
| | - Md Saidur Rahman
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh,Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, 456-756, Republic of Korea
| | - Abdullah Al Mamun Sohag
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Khandkar Shaharina Hossain
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh,Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208, Bangladesh
| | - Mithila Farjana
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh,Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208, Bangladesh
| | - Md Jamal Uddin
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh; Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
20
|
Recchiuti A, Isopi E, Romano M, Mattoscio D. Roles of Specialized Pro-Resolving Lipid Mediators in Autophagy and Inflammation. Int J Mol Sci 2020; 21:E6637. [PMID: 32927853 PMCID: PMC7555248 DOI: 10.3390/ijms21186637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic pathway that accounts for degradation and recycling of cellular components to extend cell survival under stress conditions. In addition to this prominent role, recent evidence indicates that autophagy is crucially involved in the regulation of the inflammatory response, a tightly controlled process aimed at clearing the inflammatory stimulus and restoring tissue homeostasis. To be efficient and beneficial to the host, inflammation should be controlled by a resolution program, since uncontrolled inflammation is the underlying cause of many pathologies. Resolution of inflammation is an active process mediated by a variety of mediators, including the so-called specialized pro-resolving lipid mediators (SPMs), a family of endogenous lipid autacoids known to regulate leukocyte infiltration and activities, and counterbalance cytokine production. Recently, regulation of autophagic mechanisms by these mediators has emerged, uncovering unappreciated connections between inflammation resolution and autophagy. Here, we summarize mechanisms of autophagy and resolution, focusing on the contribution of autophagy in sustaining paradigmatic examples of chronic inflammatory disorders. Then, we discuss the evidence that SPMs can restore dysregulated autophagy, hypothesizing that resolution of inflammation could represent an innovative approach to modulate autophagy and its impact on the inflammatory response.
Collapse
Affiliation(s)
| | | | | | - Domenico Mattoscio
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Sciences, University of Chieti—Pescara, 66100 Chieti, Italy; (A.R.); (E.I.); (M.R.)
| |
Collapse
|
21
|
Emamgolizadeh Gurt Tapeh B, Mosayyebi B, Samei M, Beyrampour Basmenj H, Mohammadi A, Alivand MR, Hassanpour P, Solali S. microRNAs involved in T-cell development, selection, activation, and hemostasis. J Cell Physiol 2020; 235:8461-8471. [PMID: 32324267 DOI: 10.1002/jcp.29689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) characterized by small, noncoding RNAs have a fundamental role in the regulation of gene expression at the post-transcriptional level. Additionally, miRNAs have recently been identified as potential regulators of various genes involved in the pathogenesis of the autoimmune and inflammatory disease. So far, the interaction between miRNAs and T lymphocytes in the immune response as a new and significant topic has not been emphasized substantially. The role of miRNAs in different biological processes including apoptosis, immune checkpoints and the activation of immune cells is still unclear. Aberrant miRNA expression profile affects various aspects of T-cell function. Accordingly, in this literature review, we summarized the role of significant miRNAs in T-cell development processes. Consequently, we demonstrated precise mechanisms that candidate miRNAs interfere in Immune response mediated by different types of T cells. We believe that a good understanding of the interaction between miRNAs and immune response contributes to the new therapeutic strategies in relation to disease with an immunological origin.
Collapse
Affiliation(s)
- Behnam Emamgolizadeh Gurt Tapeh
- Division of Hematology and Transfusion Medicine, Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bashir Mosayyebi
- Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Samei
- Department of Immunology, Gorgan University of Medical Sciences, Gorgan, Iran
| | | | - Ali Mohammadi
- Department of cancer and inflammation, University of Southern Denmark, Odense, Denmark
| | - Mohammad R Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Hassanpour
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Division of Hematology and Transfusion Medicine, Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Effects of an orally supplemented probiotic on the autophagy protein LC3 and Beclin1 in placentas undergoing spontaneous delivery during normal pregnancy. BMC Pregnancy Childbirth 2020; 20:216. [PMID: 32295534 PMCID: PMC7161261 DOI: 10.1186/s12884-020-02905-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Probiotic supplementation has been shown to be beneficial and is now widely promoted as an auxiliary medicine for maternal health, but the underlying mechanism is still unclear. Thus, this study aimed to explore the effects of probiotic supplementation on the placental autophagy-related proteins LC3 and Beclin1. METHOD A population-based cohort of specimens was collected under sterile conditions from 37 healthy nulliparous pregnant women who underwent systemic examination and delivered at the First Affiliated Hospital of Jinan University (Guangzhou, China). At 32 weeks of gestation, the pregnant women in the probiotic group were orally supplemented with golden bifid, and the pregnant women in the control group received no probiotic. Pregnant women with pregnancy-associated complications were excluded in the follow-up period, and 25 pregnant women undergoing spontaneous delivery were enrolled. The placental tissue specimens were collected at term. Western blotting was used to detect the protein expression, and qRT-PCR was used to detect the mRNA expression of the placental autophagy-related proteins LC3 and Beclin1. RESULTS ①There was no significant difference in the expression levels of either LC3 or Beclin1 protein between the two groups (P > 0.05). ②Probiotic supplementation induced a modest but not significant decrease in the content of LC3-mRNA with a significant decrease in the content of Beclin1-mRNA (P < 0.05). CONCLUSION Our study indicates that probiotic supplementation may reduce Beclin1-mRNA levels.
Collapse
|
23
|
Mitophagy in Acute Kidney Injury and Kidney Repair. Cells 2020; 9:cells9020338. [PMID: 32024113 PMCID: PMC7072358 DOI: 10.3390/cells9020338] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a major kidney disease characterized by rapid decline of renal function. Besides its acute consequence of high mortality, AKI has recently been recognized as an independent risk factor for chronic kidney disease (CKD). Maladaptive or incomplete repair of renal tubules after severe or episodic AKI leads to renal fibrosis and, eventually, CKD. Recent studies highlight a key role of mitochondrial pathology in AKI development and abnormal kidney repair after AKI. As such, timely elimination of damaged mitochondria in renal tubular cells represents an important quality control mechanism for cell homeostasis and survival during kidney injury and repair. Mitophagy is a selective form of autophagy that selectively removes redundant or damaged mitochondria. Here, we summarize our recent understanding on the molecular mechanisms of mitophagy, discuss the role of mitophagy in AKI development and kidney repair after AKI, and present future research directions and therapeutic potential.
Collapse
|
24
|
Bak DH, Lee E, Choi MJ, Lee BC, Kwon TR, Kim JH, Jeon ES, Oh W, Mun SK, Park BC, Na J, Kim BJ. Protective effects of human umbilical cord blood‑derived mesenchymal stem cells against dexamethasone‑induced apoptotic cell death in hair follicles. Int J Mol Med 2019; 45:556-568. [PMID: 31894311 PMCID: PMC6984800 DOI: 10.3892/ijmm.2019.4447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
Alopecia is a common and distressing condition, and developing new therapeutic agents to prevent hair loss is important. Human umbilical cord blood‑derived mesenchymal stem cells (hUCB‑MSCs) have been studied intensively in regenerative medicine. However, the therapeutic potential of these cells against hair loss and hair organ damage remains unclear, and the effects of hUCB‑MSC transplantation on hair loss require evaluation. The current study aimed to investigate the effects of hUCB‑MSCs on hair regression in vivo and restoration of anagen conduction on hair growth in vitro. The effects of hUCB‑MSCs were explored in mouse catagen induction models using a topical treatment of 0.1% dexamethasone to induce hair regression. Dexamethasone was also used to simulate a stress environment in vitro. The results demonstrated that hUCB‑MSCs significantly prevented hair regression induced by dexamethasone topical stimulation in vivo. Additionally, hUCB‑MSCs significantly increased the proliferation of human dermal papilla cells (hDPCs) and HaCaT cells, which are key constituent cells of the hair follicle. Stimulation of vascular endothelial growth factor secretion and decreased expression of DKK‑1 by hUCB‑MSCs were also observed in hDPCs. Restoration of cell viability by hUCB‑MSCs suggested that these cells exerted a protective effect on glucocorticoid stress‑associated hair loss. In addition, anti‑apoptotic effects and regulation of the autophagic flux recovery were observed in HaCaT cells. The results of the present study indicated that hUCB‑MSCs may have the capacity to protect hair follicular dermal papilla cells and keratinocytes, thus preventing hair loss. Additionally, the protective effects of hUCB‑MSCs may be resistant to dysregulation of autophagy under harmful stress.
Collapse
Affiliation(s)
- Dong Ho Bak
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, North Jeolla 56212, Republic of Korea
| | - Esther Lee
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Mi Ji Choi
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Byung Chul Lee
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Tae-Rin Kwon
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Jong-Hwan Kim
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Eun Su Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam, Gyeonggi 13494, Republic of Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam, Gyeonggi 13494, Republic of Korea
| | - Seog Kyun Mun
- Department of Otorhinolaryngology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| | - Byung Cheol Park
- Department of
Dermatology, Dankook Medical College, Cheonan, South Chungcheong 31116, Republic of Korea
| | - Jungtae Na
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| | - Beom Joon Kim
- Department of Dermatology, College of Medicine, Seoul 06973, Republic of Korea
| |
Collapse
|
25
|
Singh A, Kendall SL, Campanella M. Common Traits Spark the Mitophagy/Xenophagy Interplay. Front Physiol 2018; 9:1172. [PMID: 30294276 PMCID: PMC6158333 DOI: 10.3389/fphys.2018.01172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/03/2018] [Indexed: 12/16/2022] Open
Abstract
Selective autophagy contributes to the wellbeing of eukaryotic cells by recycling cellular components, disposing damaged organelles, and removing pathogens, amongst others. Both the quality control process of selective mitochondrial autophagy (Mitophagy) and the defensive process of intracellular pathogen-engulfment (Xenophagy) are facilitated via protein assemblies which have shared molecules, a prime example being the Tank-Binding Kinase 1 (TBK1). TBK1 plays a central role in the immunity response driven by Xenophagy and was recently shown to be an amplifying mechanism in Mitophagy, bring to attention the potential cross talk between the two processes. Here we draw parallels between Xenophagy and Mitophagy, speculating on the inhibitory mechanisms of specific proteins (e.g., the 18 kDa protein TSPO), how the preferential sequestering toward one of the two pathways may undermine the other, and in this way impair cellular response to pathogens and cellular immunity. We believe that an in depth understanding of the commonalities may present an opportunity to design novel therapeutic strategies targeted at both the autonomous and non-autonomous processes of selective autophagy.
Collapse
Affiliation(s)
- Aarti Singh
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Sharon L Kendall
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hertfordshire, United Kingdom
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,UCL Consortium for Mitochondrial Research, London, United Kingdom
| |
Collapse
|
26
|
Polymorphisms in Autophagy-Related Gene IRGM Are Associated with Susceptibility to Autoimmune Thyroid Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7959707. [PMID: 29992164 PMCID: PMC6016217 DOI: 10.1155/2018/7959707] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/21/2018] [Accepted: 03/26/2018] [Indexed: 01/19/2023]
Abstract
Background To date, studies have shown that polymorphisms in an autophagy-related gene, IRGM, are linked with different diseases, especially autoimmune diseases. The present study aimed to examine the roles of IRGM polymorphisms in autoimmune thyroid diseases (AITD). Methods Three polymorphisms in IRGM gene (rs10065172, rs4958847, and rs13361189) were genotyped in 1569 participants (488 with Graves' disease, 292 with Hashimoto's thyroiditis, and 789 healthy controls) using PCR-based ligase detection reaction method. Gene-disease associations were evaluated for the three SNPs. Results T allele of rs10065172, A allele of rs4958847, and C allele of rs13361189 were all higher in Graves' disease patients than controls, and the ORs were OR = 1.207 (P = 0.022), OR = 1.207 (P = 0.027), and OR = 1.200 (P = 0.027), respectively. After adjusting for sex and age, rs10065172 and rs13361189 were still associated with GD under both the allele model and dominant model, and the adjusted ORs for rs10065172 were 1.20 (P = 0.033) and 1.33 (P = 0.024), while the adjusted ORs for rs13361189 were 1.19 (P = 0.042) and 1.33 (P = 0.026), respectively. No significant difference was found between Hashimoto's thyroiditis patients and controls. Haplotype analysis found that CTA frequency was distinguishingly higher in Graves' disease patients (OR = 1.195, P = 0.030). The frequency of TCG haplotype was distinguishingly lower in AITD and Graves' disease patients (OR = 0.861, P = 0.044; OR = 0.816, P = 0.017). Conclusions Our study reveals IRGM as a susceptibility gene of AITD and Graves' disease for the first time.
Collapse
|
27
|
Abele R, Tampé R. Moving the Cellular Peptidome by Transporters. Front Cell Dev Biol 2018; 6:43. [PMID: 29761100 PMCID: PMC5937356 DOI: 10.3389/fcell.2018.00043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/03/2018] [Indexed: 12/18/2022] Open
Abstract
Living matter is defined by metastability, implying a tightly balanced synthesis and turnover of cellular components. The first step of eukaryotic protein degradation via the ubiquitin-proteasome system (UPS) leads to peptides, which are subsequently degraded to single amino acids by an armada of proteases. A small fraction of peptides, however, escapes further cytosolic destruction and is transported by ATP-binding cassette (ABC) transporters into the endoplasmic reticulum (ER) and lysosomes. The ER-resident heterodimeric transporter associated with antigen processing (TAP) is a crucial component in adaptive immunity for the transport and loading of peptides onto major histocompatibility complex class I (MHC I) molecules. Although the function of the lysosomal resident homodimeric TAPL-like (TAPL) remains, until today, only loosely defined, an involvement in immune defense is anticipated since it is highly expressed in dendritic cells and macrophages. Here, we compare the gene organization and the function of single domains of both peptide transporters. We highlight the structural organization, the modes of substrate binding and translocation as well as physiological functions of both organellar transporters.
Collapse
Affiliation(s)
- Rupert Abele
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany.,Cluster of Excellence - Macromolecular Complexes, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
28
|
Autophagy-associated immune responses and cancer immunotherapy. Oncotarget 2018; 7:21235-46. [PMID: 26788909 PMCID: PMC5008281 DOI: 10.18632/oncotarget.6908] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/06/2016] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved catabolic process by which cellular components are sequestered into a double-membrane vesicle and delivered to the lysosome for terminal degradation and recycling. Accumulating evidence suggests that autophagy plays a critical role in cell survival, senescence and homeostasis, and its dysregulation is associated with a variety of diseases including cancer, cardiovascular disease, neurodegeneration. Recent studies show that autophagy is also an important regulator of cell immune response. However, the mechanism by which autophagy regulates tumor immune responses remains elusive. In this review, we will describe the role of autophagy in immune regulation and summarize the possible molecular mechanisms that are currently well documented in the ability of autophagy to control cell immune response. In addition, the scientific and clinical hurdles regarding the potential role of autophagy in cancer immunotherapy will be discussed.
Collapse
|
29
|
Li L, Jin H, Wang H, Cao Z, Feng N, Wang J, Zhao Y, Zheng X, Hou P, Li N, Chi H, Huang P, Jiao C, Li Q, Wang L, Wang T, Sun W, Gao Y, Tu C, Hu G, Yang S, Xia X. Autophagy is highly targeted among host comparative proteomes during infection with different virulent RABV strains. Oncotarget 2017; 8:21336-21350. [PMID: 28186992 PMCID: PMC5400588 DOI: 10.18632/oncotarget.15184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Rabies virus (RABV) is a neurotropic virus that causes serious disease in humans and animals worldwide. It has been reported that different RABV strains can result in divergent prognoses in animal model. To identify host factors that affect different infection processes, a kinetic analysis of host proteome alterations in mouse brains infected with different virulent RABV strains was performed using isobaric tags for a relative and absolute quantification (iTRAQ)-liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomics approach, and this analysis identified 147 differentially expressed proteins (DEPs) between the pathogenic challenge virus standard (CVS)-11 strain and the attenuated SRV9 strain. Bioinformatics analyses of these DEPs revealed that autophagy and several pathways associated with autophagy, such as mammalian target of rapamycin (mTOR) signaling, p70S6K signaling, nuclear factor erythroid 2-related factor 2 (NRF2)-mediated oxidative stress and superoxide radical degradation, were dysregulated. Validation of the proteomic data showed that attenuated SRV9 induced more autophagosome accumulation than CVS-11 in an in vitro model. Our findings provide new insights into the pathogenesis of RABV and encourage further studies on this topic.
Collapse
Affiliation(s)
- Ling Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Hongli Jin
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Zengguo Cao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Jianzhong Wang
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Xuexing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,School of Public Health, Shandong University, Jinan, China
| | - Pengfei Hou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Hang Chi
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Pei Huang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Cuicui Jiao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Qian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Lina Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Weiyang Sun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Changchun Tu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Guixue Hu
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| |
Collapse
|
30
|
Abstract
Unlike B cells, CD8-positive and CD4-positive T cells of the adaptive immune system do not recognize intact foreign proteins but instead recognize polypeptide fragments of potential antigens. These antigenic peptides are expressed on the surface of antigen presenting cells bound to MHC class I and MHC class II proteins. Here, we review the basics of antigen acquisition by antigen presenting cells, antigen proteolysis into polypeptide fragments, antigenic peptide binding to MHC proteins, and surface display of both MHC class I-peptide and MHC class II-peptide complexes.
Collapse
|
31
|
Ochi A, Chen D, Schulte W, Leng L, Moeckel N, Piecychna M, Averdunk L, Stoppe C, Bucala R, Moeckel G. MIF-2/D-DT enhances proximal tubular cell regeneration through SLPI- and ATF4-dependent mechanisms. Am J Physiol Renal Physiol 2017; 313:F767-F780. [PMID: 28539339 DOI: 10.1152/ajprenal.00683.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine with pleiotropic actions that is produced by several organs and cell types. Depending on the target cell and the inflammatory context, MIF can engage its two component receptor complex CD74 and CD44 and the chemokine receptors CXCR2/4. MIF is constitutively expressed in renal proximal tubular cells, stored in intracellular preformed pools, and released at a low rate. Recently, a second MIF-like protein (i.e., MIF-2/D-DT) has been characterized in mammals. Our study was aimed at examining the role of MIF-2/D-DT, which mediates tissue protection in the heart, in tubular cell regeneration from ischemia-reperfusion injury. We found that Mif-/-, Mif-2-/-, and Cd74-/- mice had significantly worse tubular injury compared with wild-type (WT) control mice and that treatment with MIF-2/D-DT significantly improved recovery of injured epithelial cells. RNAseq analysis of kidney tissue from the ischemia-reperfusion injury model revealed that MIF-2/D-DT treatment stimulates secretory leukocyte proteinase inhibitor (SLPI) and cyclin D1 expression. MIF-2/D-DT additionally activates of eukaryotic initiation factor (eIF) 2α and activating transcription factor (ATF) 4, two transcription factors involved in the integrated stress response (ISR), which is a cellular stress response activated by hypoxia, nutrient deprivation, and oxygen radicals. MIF-2/D-DT also inhibited apoptosis and induced autophagy in hypoxia-treated mouse proximal tubular (MPT) cells. These results indicate that MIF-2/D-DT is an important factor in tubular cell regeneration and may be of therapeutic utility as a regenerative agent in the clinical setting of ischemic acute kidney injury.
Collapse
Affiliation(s)
- Akinobu Ochi
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Dong Chen
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Wibke Schulte
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Nickolas Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Marta Piecychna
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Luisa Averdunk
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.,Department of Anesthesiology and Intensive Care Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Christian Stoppe
- Department of Anesthesiology and Intensive Care Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut;
| |
Collapse
|
32
|
Abstract
The protective effect of meningococcal vaccines targeting disease causing serogroups exemplified by the introduction of MenAfriVac™ in Africa, is well established and documented in large population-based studies. Due to the emergence of other meningococcal disease causing serogroups, novel vaccine formulations are needed. There is a high potential for novel nanotechnology-based meningococcal vaccine formulations that can provide wider vaccine coverage. The proposed meningococcal vaccine formulation contains spherical shaped micro and nanoparticles that are biological mimics of Niesseria meningitidis, therefore present to immune system as invader and elicit robust immune responses. Vaccine nanoparticles encapsulate meningococcal CPS polymers in a biodegradable material that slowly release antigens, therefore enhance antigen presentation by exerting antigen depot effect. The antigenicity of meningococcal vaccine delivered in nanoparticles is significantly higher when compared to vaccine delivered in solution. Preclinical studies are required to assess the immunogenicity of novel vaccine formulations. Therefore, implementing various in-vitro human immune cell-based assays that mimic in-vivo interactions, would provide good insight on optimal antigen dose, effective antigen presentation, facilitate screening of various vaccine and adjuvant combinations and predict in-vivo immunogenicity. This rapid approach is cost-effective and provides data required for the preclinical immunogenicity assessment of novel meningococcal vaccine formulations.
Collapse
Affiliation(s)
- Susu M Zughaier
- a Laboratory of Bacterial Pathogenesis , Department of Veterans Affairs Medical Center , Decatur , GA , USA.,b Department of Microbiology and Immunology , Emory University School of Medicine , Atlanta , GA , USA
| |
Collapse
|
33
|
Hu L, Jiang K, Ding C, Meng S. Targeting Autophagy for Oncolytic Immunotherapy. Biomedicines 2017; 5:biomedicines5010005. [PMID: 28536348 PMCID: PMC5423490 DOI: 10.3390/biomedicines5010005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OVs) are capable of exerting anti-cancer effects by a variety of mechanisms, including immune-mediated tumor cell death, highlighting their potential use in immunotherapy. Several adaptation mechanisms such as autophagy contribute to OV-mediated anti-tumor properties. Autophagy regulates immunogenic signaling during cancer therapy which can be utilized to design therapeutic combinations using approaches that either induce or block autophagy to potentiate the therapeutic efficacy of OVs. In this article, we review the complicated interplay between autophagy, cancer, immunity, and OV, summarize recent progress in the contribution of OV-perturbed autophagy to oncolytic immunity, and discuss the challenges in targeting autophagy to enhance oncolytic immunotherapy.
Collapse
Affiliation(s)
- Lulu Hu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| | - Ke Jiang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200000, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| |
Collapse
|
34
|
van Niekerk G, Loos B, Nell T, Engelbrecht AM. Autophagy--A free meal in sickness-associated anorexia. Autophagy 2016; 12:727-34. [PMID: 27050464 DOI: 10.1080/15548627.2016.1147672] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Activation of the immune system is metabolically costly, yet a hallmark of an infection is a reduction in appetite with a subsequent reduction in metabolite provision. What is the functional value of decreasing nutrient intake when an infection imposes large demands on metabolic parameters? Here, we propose that sickness-associated anorexia (SAA) upregulates the ancient process of autophagy systemically, thereby profoundly controlling not only immune- but also nonimmune-competent cells. This allows an advanced impact on the resolution of an infection through direct pathogen killing, enhancement of epitope presentation and the contribution toward the clearance of noxious factors. By rendering a 'free meal,' autophagy is thus most fundamentally harnessed during an anorexic response in order to promote both host tolerance and resistance. These findings strongly suggest a reassessment of numerous SAA-related clinical applications and a re-evaluation of current efforts in patient care.
Collapse
Affiliation(s)
- Gustav van Niekerk
- a Department of Physiological Sciences , Stellenbosch University , Stellenbosch , South Africa
| | - Ben Loos
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| | - Theo Nell
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| | - Anna-Mart Engelbrecht
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| |
Collapse
|
35
|
Martin C, Espaillat MP, Santiago-Schwarz F. IL-10 restricts dendritic cell (DC) growth at the monocyte-to-monocyte-derived DC interface by disrupting anti-apoptotic and cytoprotective autophagic molecular machinery. Immunol Res 2016; 63:131-43. [PMID: 26395023 DOI: 10.1007/s12026-015-8700-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An evolving premise is that cytoprotective autophagy responses are essential to monocyte-macrophage differentiation. Whether autophagy functions similarly during the monocyte-to-dendritic cell (DC) transition is unclear. IL-10, which induces apoptosis in maturing human DCs, has been shown to inhibit starvation-induced autophagy in murine macrophage cell lines. Based on the strict requirement that Bcl-2-mediated anti-apoptotic processes are implemented during the monocyte-to-DC transition, we hypothesized that cytoprotective autophagy responses also operate at the monocyte-DC interface and that IL-10 inhibits both anti-apoptotic and cytoprotective autophagy responses at this critical juncture. In support of our premise, we show that levels of anti-apoptotic Bcl-2 and autophagy-associated LC3 and Beclin-1 proteins are coincidentally upregulated during the monocyte-to-DC transition. Autophagy was substantiated by increased autophagosome visualization after bafilomycin treatment. Moreover, the autophagy inhibitor 3-MA restricted DC differentiation by prompting apoptosis. IL-10 implemented apoptosis that was coincidentally associated with reduced levels of Bcl-2 and widespread disruption of the autophagic flux. During peak apoptosis, IL-10 produced the death of newly committed DCs. However, cells surviving the IL-10 apoptotic schedule were highly phagocytic macrophage-like cells displaying reduced capacity to stimulate allogeneic naïve T cells in a mixed leukocyte reaction, increased levels of LC3, and mature autophagosomes. Thus, IL-10's negative control of DC-driven adaptive immunity at the monocyte-DC interface includes disruption of coordinately regulated molecular networks involved in pro-survival autophagy and anti-apoptotic responses.
Collapse
Affiliation(s)
- Carla Martin
- Biology Department, Farmingdale State College, State University of New York, Farmingdale, NY, 11735, USA
| | - Mel Pilar Espaillat
- Molecular Genetics and Microbiology and Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY, 11794, USA
| | - Frances Santiago-Schwarz
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
36
|
Pei J, Deng J, Ye Z, Wang J, Gou H, Liu W, Zhao M, Liao M, Yi L, Chen J. Absence of autophagy promotes apoptosis by modulating the ROS-dependent RLR signaling pathway in classical swine fever virus-infected cells. Autophagy 2016; 12:1738-1758. [PMID: 27463126 DOI: 10.1080/15548627.2016.1196318] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A growing number of studies have demonstrated that both macroautophagy/autophagy and apoptosis are important inner mechanisms of cell to maintain homeostasis and participate in the host response to pathogens. We have previously reported that a functional autophagy pathway is trigged by infection of classical swine fever virus (CSFV) and is required for viral replication and release in host cells. However, the interplay of autophagy and apoptosis in CSFV-infected cells has not been clarified. In the present study, we demonstrated that autophagy induction with rapamycin facilitates cellular proliferation after CSFV infection, and that autophagy inhibition by knockdown of essential autophagic proteins BECN1/Beclin 1 or MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) promotes apoptosis via fully activating both intrinsic and extrinsic mechanisms in CSFV-infected cells. We also found that RIG-I-like receptor (RLR) signaling was amplified in autophagy-deficient cells during CSFV infection, which was closely linked to the activation of the intrinsic apoptosis pathway. Moreover, we discovered that virus infection of autophagy-impaired cells results in an increase in copy number of mitochondrial DNA and in the production of reactive oxygen species (ROS), which plays a significant role in enhanced RLR signaling and the activated extrinsic apoptosis pathway in cultured cells. Collectively, these data indicate that CSFV-induced autophagy delays apoptosis by downregulating ROS-dependent RLR signaling and thus contributes to virus persistent infection in host cells.
Collapse
Affiliation(s)
- Jingjing Pei
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jieru Deng
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Zuodong Ye
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jiaying Wang
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Hongchao Gou
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Wenjun Liu
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Mingqiu Zhao
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Ming Liao
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Lin Yi
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jinding Chen
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| |
Collapse
|
37
|
Mao S, Zhang J. Role of autophagy in chronic kidney diseases. Int J Clin Exp Med 2015; 8:22022-9. [PMID: 26885176 PMCID: PMC4729962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 12/06/2015] [Indexed: 06/05/2023]
Abstract
Chronic kidney diseases (CKD), a common pathway of various glomerular diseases, which carries great morbidity and mortality to people. CKD is characterized by progressive kidney fibrosis and remodeling. CKD is also associated with the depletion of glomerular and tubular cells. Autophagy is a highly conserved process that degrades cellular long-lived proteins and organelles. It plays an important role in both normal and disease states, including immunity, inflammation, and adaptation to stress. Evidence has indicated that impaired autophagic activity is involved in the development of CKD. Here, we review the progress in our understanding of the role of autophagy in the development and progression of CKD. Targeting the autophagic signaling pathways may be a therapeutic strategy for CKD.
Collapse
Affiliation(s)
- Song Mao
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Jianhua Zhang
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| |
Collapse
|
38
|
Duraes FV, Niven J, Dubrot J, Hugues S, Gannagé M. Macroautophagy in Endogenous Processing of Self- and Pathogen-Derived Antigens for MHC Class II Presentation. Front Immunol 2015; 6:459. [PMID: 26441964 PMCID: PMC4585038 DOI: 10.3389/fimmu.2015.00459] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022] Open
Abstract
Although autophagy is a process that has been studied for several years its link with antigen presentation and T cell immunity has only recently emerged. Autophagy, which means “self-eating,” is important to maintain cell homeostasis and refers to a collection of mechanisms that delivers intracellular material for degradation into lysosomes. Among them, macroautophagy pathway has many implications in different biological processes, including innate and adaptive immunity. In particular, macroautophagy can provide a substantial source of intracellular antigens for loading onto MHC class II molecules using the alternative MHC class II pathway. Through autophagosomes, endogenous self-antigens as well as antigens derived from intracellular pathogens can be delivered to MHC class II compartment and presented to CD4+ T cells. The pathway will, therefore, impact both peripheral T cell tolerance and the pathogen specific immune response. This review will describe the contribution of autophagy to intracellular presentation of endogenous self- or pathogen-derived antigens via MHC class II and its consequences on CD4+ T cell responses.
Collapse
Affiliation(s)
- Fernanda V Duraes
- Department of Pathology and Immunology, School of Medicine, University of Geneva , Geneva , Switzerland
| | - Jennifer Niven
- Department of Pathology and Immunology, School of Medicine, University of Geneva , Geneva , Switzerland ; Division of Rheumatology, Department of Internal Medicine, University Hospital Geneva , Geneva , Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, School of Medicine, University of Geneva , Geneva , Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, School of Medicine, University of Geneva , Geneva , Switzerland
| | - Monique Gannagé
- Department of Pathology and Immunology, School of Medicine, University of Geneva , Geneva , Switzerland ; Division of Rheumatology, Department of Internal Medicine, University Hospital Geneva , Geneva , Switzerland
| |
Collapse
|
39
|
Oliveira CC, van Hall T. Alternative Antigen Processing for MHC Class I: Multiple Roads Lead to Rome. Front Immunol 2015; 6:298. [PMID: 26097483 PMCID: PMC4457021 DOI: 10.3389/fimmu.2015.00298] [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: 03/31/2015] [Accepted: 05/22/2015] [Indexed: 12/31/2022] Open
Abstract
The well described conventional antigen-processing pathway is accountable for most peptides that end up in MHC class I molecules at the cell surface. These peptides experienced liberation by the proteasome and transport by the peptide transporter TAP. However, there are multiple roads that lead to Rome, illustrated by the increasing number of alternative processing pathways that have been reported during last years. Interestingly, TAP-deficient individuals do not succumb to viral infections, suggesting that CD8 T cell immunity is sufficiently supported by alternative TAP-independent processing pathways. To date, a diversity of viral and endogenous TAP-independent peptides have been identified in the grooves of different MHC class I alleles. Some of these peptides are not displayed by normal TAP-positive cells and we therefore called them TEIPP, for “T-cell epitopes associated with impaired peptide processing.” TEIPPs are hidden self-antigens, are derived from normal housekeeping proteins, and are processed via unconventional processing pathways. Per definition, TEIPPs are presented via TAP-independent pathways, but recent data suggest that part of this repertoire still depend on proteasome and metalloprotease activity. An exception is the C-terminal peptide of the endoplasmic reticulum (ER)-membrane-spanning ceramide synthase Trh4 that is surprisingly liberated by the signal peptide peptidase (SPP), the proteolytic enzyme involved in cleaving leader sequences. The intramembrane cleaving SPP is thereby an important contributor of TAP-independent peptides. Its family members, like the Alzheimer’s related presenilins, might contribute as well, according to our preliminary data. Finally, alternative peptide routing is an emerging field and includes processes like the unfolded protein response, the ER-associated degradation, and autophagy-associated vesicular pathways. These data convince us that there is a world to be discovered in the field of unconventional antigen processing.
Collapse
Affiliation(s)
- Cláudia C Oliveira
- Department of Clinical Oncology, Leiden University Medical Center , Leiden , Netherlands
| | - Thorbald van Hall
- Department of Clinical Oncology, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
40
|
The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 2015; 15:203-16. [PMID: 25720354 DOI: 10.1038/nri3818] [Citation(s) in RCA: 730] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antigenic peptide-loaded MHC class II molecules (peptide-MHC class II) are constitutively expressed on the surface of professional antigen-presenting cells (APCs), including dendritic cells, B cells, macrophages and thymic epithelial cells, and are presented to antigen-specific CD4(+) T cells. The mechanisms of antigen uptake, the nature of the antigen processing compartments and the lifetime of cell surface peptide-MHC class II complexes can vary depending on the type of APC. It is likely that these differences are important for the function of each distinct APC subset in the generation of effective adaptive immune responses. In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide-MHC class II complexes, the intracellular trafficking of peptide-MHC class II complexes to the APC plasma membrane and their ultimate degradation.
Collapse
|
41
|
Abstract
The autophagy pathway represents an evolutionarily conserved cell recycling process that is activated in response to nutrient deprivation and other stress signals. Over the years, it has been linked to an array of cellular functions. Equally, a wide range of cell-intrinsic, as well as extracellular, factors have been implicated in the induction of the autophagy pathway. Microbial infections represent one such factor that can not only activate autophagy through specific mechanisms but also manipulate the response to the invading microbe’s advantage. Moreover, in many cases, particularly among viruses, the pathway has been shown to be intricately involved in the replication cycle of the pathogen. Conversely, autophagy also plays a role in combating the infection process, both through direct destruction of the pathogen and as one of the key mediating factors in the host defense mechanisms of innate and adaptive immunity. Further, the pathway also plays a role in controlling the pathogenesis of infectious diseases by regulating inflammation. In this review, we discuss various interactions between pathogens and the cellular autophagic response and summarize the immunological functions of the autophagy pathway.
Collapse
Affiliation(s)
- Mayura Desai
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Rong Fang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|
42
|
Meiners S, Ballweg K. Proteostasis in pediatric pulmonary pathology. Mol Cell Pediatr 2014; 1:11. [PMID: 26567105 PMCID: PMC4530569 DOI: 10.1186/s40348-014-0011-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023] Open
Abstract
Protein homeostasis describes the tight supervision of protein synthesis, correct protein maturation and folding, as well as the timely disposal of unwanted and damaged proteins by the ubiquitin-proteasome pathway or the lysosome-autophagy route. The cellular processes involved in preservation of protein homeostasis are collectively called proteostasis. Dysregulation of proteostasis is an emerging common pathomechanism for chronic lung diseases in the adult and aged patient. There is also rising evidence that impairment of protein homeostasis contributes to early sporadic disease onset in pediatric lung diseases beyond the well-known hereditary proteostasis disorders such as cystic fibrosis and alpha-1 antitrypsin (AAT) deficiency. Identifying the pathways that contribute to impaired proteostasis will provide new avenues for therapeutic interference with the pathogenesis of chronic lung diseases in the young and adult. Here, we introduce the concept of proteostasis and summarize available evidence on dysregulation of proteostasis pathways in pediatric and adult chronic lung diseases.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians-Universität, Asklepios Klinik Gauting und Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377, Munich, Germany.
| | - Korbinian Ballweg
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians-Universität, Asklepios Klinik Gauting und Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377, Munich, Germany.
| |
Collapse
|
43
|
Vujanovic L, Shi J, Kirkwood JM, Storkus WJ, Butterfield LH. Molecular mimicry of MAGE-A6 and Mycoplasma penetrans HF-2 epitopes in the induction of antitumor CD8 + T-cell responses. Oncoimmunology 2014; 3:e954501. [PMID: 25960935 DOI: 10.4161/21624011.2014.954501] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/11/2014] [Indexed: 11/19/2022] Open
Abstract
A promising vaccine strategy for the treatment of cancer involves the use of vaccines incorporating tumor antigen-derived synthetic peptides that can be coordinately recognized by specific CD4+ and CD8+ T-cells. Previously, we reported that a MAGE-A6-derived peptide (MAGE-A6172-187) and its highly-immunogenic and cross-reactive homolog derived from Mycoplasma penetrans HF-2 permease (HF-2216-229) are promiscuously presented by multiple HLA-DR alleles to responder CD4+ T-cells obtained from healthy donors and melanoma patients. Here, we investigated whether these same peptides could concomitantly stimulate cross-reactive MAGE-A6-specific CD8+ T-cell responses in vitro using cells isolated from HLA-A*0201 (HLA-A2)+ healthy individuals and patients with melanoma. We now show that MAGE-A6172-187 and, even more so, HF-2216-229, induce memory CD8+ T cells that recognize HLA-A2+ MAGE-A6+ tumor target cells. The immunogenicity of these peptides was at least partially attributed to their embedded MAGE-A6176-185 and HF-2220-229 "homologous" sequences. The functional avidity of HF-2216-229 peptide-primed CD8+ T cells for the MAGE-A6172-187 peptide was more than 100-fold greater than that of CD8+ T cells primed with the corresponding MAGE-A6 peptide. Additionally, these 2 peptides were recognized in interferon γ (IFNγ) and granzyme B ELISPOT assays by CD8+ T-cell clones displaying variable T-cell receptor (TCR) Vβ usage. These data suggest that the immune cross-reactivity of the MAGE-A6172-187 and HF-2216-229 peptides extends to CD8+ T cells, at least in HLA-A2+ donors, and supports the potential translational utility of these epitopes in clinical vaccine formulations and for immunomonitoring of cancer patients.
Collapse
Key Words
- APC, antigen presenting cell
- AdV, recombinant adenoviral vector
- CD8+ T-cell
- CTL, cytotoxic T lymphocyte
- EBV, Epstein-Barr virus
- FBS, fetal bovine serum
- HD, healthy donor
- HLA, human leukocyte antigen
- HPLC, high-performance liquid chromatography
- IVS, in vitro stimulation
- MACS, Magnetic-Activated Cell Sorting
- MAGE-A6
- MOI, multiplicity of infection
- Mycoplasma penetrans
- PBMC, peripheral blood mononuclear cell
- PFU, plaque forming units
- RT-PCR, reverse transcription polymerase chain reaction
- TAA, tumor associated antigen
- TCM, T cell media
- TCR, T-cell receptor
- epitope mimic
- iDC, immature dendritic cells
- mDC, mature dendritic cells
- melanoma
Collapse
Affiliation(s)
- Lazar Vujanovic
- University of Pittsburgh Cancer Institute ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Medicine ; Pittsburgh, PA USA
| | - Jian Shi
- University of Pittsburgh Cancer Institute ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Medicine ; Pittsburgh, PA USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Medicine ; Pittsburgh, PA USA
| | - Walter J Storkus
- University of Pittsburgh Cancer Institute ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Immunology ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Dermatology ; Pittsburgh, PA USA
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Medicine ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Immunology ; Pittsburgh, PA USA ; University of Pittsburgh School of Medicine; Department of Surgery ; Pittsburgh, PA USA
| |
Collapse
|
44
|
He L, Livingston MJ, Dong Z. Autophagy in acute kidney injury and repair. Nephron Clin Pract 2014; 127:56-60. [PMID: 25343822 DOI: 10.1159/000363677] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Acute kidney injury (AKI) is a major kidney disease associated with a poor clinical outcome both in the short and long term. Autophagy is a cellular stress response that plays important roles in the pathogenesis of various diseases. Autophagy is induced in proximal tubules during AKI. A renoprotective role of autophagy in AKI has been demonstrated by pharmacological and genetic inhibition studies. The role of autophagy in kidney recovery and repair from AKI, however, remains largely unknown. A dynamic change in autophagy during the recovery phase of AKI seems to be important for tubular proliferation and repair. In renal fibrosis, autophagy may either promote this via the induction of tubular atrophy and decomposition, or prevent it via effects on the intracellular degradation of excessive collagen. Further research is expected to improve the understanding of the regulation of autophagy in kidney injury and repair, elucidate the pathological roles of autophagy in renal fibrosis, and discover therapeutic targets for treating AKI and preventing its progression to chronic kidney disease.
Collapse
Affiliation(s)
- Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, PR China
| | | | | |
Collapse
|
45
|
Gros F, Muller S. Pharmacological regulators of autophagy and their link with modulators of lupus disease. Br J Pharmacol 2014; 171:4337-59. [PMID: 24902607 DOI: 10.1111/bph.12792] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/23/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a central regulator of cell survival. It displays both anti- and pro-death roles that are decisive in the maintenance of cell homeostasis. Initially described in several eukaryotic cellular models as being induced under nutrient stress favouring survival by energy supply, autophagy was found later to display other decisive physiological roles, especially in the immune system. Thus, it is involved in antigen presentation and lymphocyte differentiation as well as in the balance regulating survival/death and activation of lymphocytes. Autophagy therefore appears to be central in the regulation of inflammation. The observation that autophagy is deregulated in systemic lupus erythematosus is recent. This discovery revives the programme dealing with the design and development of pharmacological autophagy regulators in the therapeutic context of lupus, a debilitating autoimmune disease that affects several million people in the world. A large number of molecules that positively and negatively regulate autophagy have been described, most of them with therapeutic indications in cancer and infection. Only a few, however, are effectively potent activators or inhibitors endowed with experimentally demonstrated selective properties. In this review article, we highlight the most relevant ones and summarize what we know regarding their mechanism of action. We emphasize the link between pharmacological regulators of autophagy and inducers or inhibitors of lupus disease and discuss the fundamental and pharmacological/therapeutic interest of this functional interplay.
Collapse
Affiliation(s)
- Frédéric Gros
- CNRS, Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France; University of Strasbourg, Strasbourg, France
| | | |
Collapse
|
46
|
Abstract
Plasma cells (PCs) are the effectors responsible for antibody (Ab)-mediated immunity. They differentiate from B lymphocytes through a complete remodeling of their original structure and function. Stress is a constitutive element of PC differentiation. Macroautophagy, conventionally referred to as autophagy, is a conserved lysosomal recycling strategy that integrates cellular metabolism and enables adaptation to stress. In metazoa, autophagy plays diverse roles in cell differentiation. Recently, a number of autophagic functions have been recognized in innate and adaptive immunity, including clearance of intracellular pathogens, inflammasome regulation, lymphocyte ontogenesis, and antigen presentation. We identified a previously unrecognized role played by autophagy in PC differentiation and activity. Following B cell activation, autophagy moderates the expression of the transcriptional repressor Blimp-1 and immunoglobulins through a selective negative control exerted on the size of the endoplasmic reticulum and its stress signaling response, including the essential PC transcription factor, XBP-1. This containment of PC differentiation and function, i.e., Ab production, is essential to optimize energy metabolism and viability. As a result, autophagy sustains Ab responses in vivo. Moreover, autophagy is an essential intrinsic determinant of long-lived PCs in their as yet poorly understood bone marrow niche. In this essay, we discuss these findings in the context of the established biological functions of autophagy, and their manifold implications for adaptive immunity and PC diseases, in primis multiple myeloma.
Collapse
Affiliation(s)
- Laura Oliva
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy
| | - Simone Cenci
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy ; Università Vita-Salute San Raffaele , Milan , Italy ; Bone Pathophysiology Program (BoNetwork), Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy
| |
Collapse
|
47
|
Zullo AJ, Jurcic Smith KL, Lee S. Mammalian target of Rapamycin inhibition and mycobacterial survival are uncoupled in murine macrophages. BMC BIOCHEMISTRY 2014; 15:4. [PMID: 24528777 PMCID: PMC3937017 DOI: 10.1186/1471-2091-15-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/10/2014] [Indexed: 01/06/2023]
Abstract
Background Autophagy is a cellular response to intracellular pathogens including mycobacteria and is induced by the direct inhibitors of mammalian target of Rapamycin (mTOR), a major negative regulator of autophagy. Autophagy induction by mTOR inhibition (mTOR dependent autophagy), through chemical means or starvation, leads to mycobacterial killing in infected cells. However, previous work by our group has shown that mycobacterial infection of macrophages naturally induces both autophagy and mammalian target of Rapamycin (mTOR) activity (mTOR independent autophagy). In the current work, we further explore the relationship between mTOR activity and mycobacterial killing in macrophages. Results While low concentrations of the mTOR inhibitors, Rapamycin, Torin 1, and Torin 2, can effectively reduce or block mTOR activity in response to lipopolysaccharides (LPS) or mycobacteria, higher concentrations (10 uM) are required to observe Mycobacterium smegmatis killing. The growth of M. smegmatis was also inhibited by high concentrations of Rapamycin in LC3B and ATG5 deficient bone marrow derived macrophages, suggesting that non-autophagic mechanisms might contribute to killing at high doses. Since mycobacterial killing could be observed only at fairly high concentrations of the mTOR inhibitors, exceeding doses necessary to inhibit mTOR, we hypothesized that high doses of Rapamycin, the most commonly utilized mTOR inhibitor for inducing autophagic killing, may exert a direct bactericidal effect on the mycobacteria. Although a short-term treatment of mycobacteria with Rapamycin did not substantially affect mycobacterial growth, a long-term exposure to Rapamycin could impact mycobacterial growth in vitro in select species. Conclusions This data, coupled with previous work from our laboratory, further indicates that autophagy induction by mTOR inhibition is an artificial means to increase mycobacterial killing and masks more relevant endogenous autophagic biochemistry that needs to be understood.
Collapse
Affiliation(s)
| | | | - Sunhee Lee
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
48
|
Dalton JP, Desmond A, Shanahan F, Hill C. Detection of Mycobacterium avium subspecies paratuberculosis in patients with Crohn's disease is unrelated to the presence of single nucleotide polymorphisms rs2241880 (ATG16L1) and rs10045431 (IL12B). Med Microbiol Immunol 2014; 203:195-205. [PMID: 24522266 DOI: 10.1007/s00430-014-0332-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/31/2014] [Indexed: 12/23/2022]
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) has been controversially linked with Crohn's disease (CD). Detection of MAP in CD has been highly variable, and one explanation might be the genetic heterogeneity of this syndrome. Many of the single nucleotide polymorphisms (SNPs) linked with CD are contained within genes that are associated with bacterial handling in general, and some are specifically implicated in susceptibility to mycobacterial disease. We tested a cohort of IBD patients (n = 149) to determine whether the presence of MAP was associated with a selection of these SNPs. Blood samples from CD patients (n = 84), ulcerative colitis (UC, n = 65) patients and healthy controls (n = 55) were examined for the presence of MAP and SNPs in ATG16L1, IL12B, NOD2/CARD15, NKx2-3, IL23R and IRGM. Statistical analysis was then used to determine whether there was any association between the presence of MAP and these SNPs. MAP, rs2241880 (ATG16L1) and rs10045431 (IL12B) were found to be significantly associated with CD. The presence of MAP was not related to the status of the SNPs in ATG16L1 or IL12B. We have found no evidence for the contribution of these SNPs to the presence of MAP in CD patients.
Collapse
Affiliation(s)
- James P Dalton
- Department of Microbiology, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
49
|
Wang L, Song J, Zhang J, Zhu C, Ma Y, Xu X. Lentiviral vector-mediate ATG3 overexpression inhibits growth and promotes apoptosis of human SKM-1 cells. Mol Biol Rep 2014; 41:2093-9. [PMID: 24420857 DOI: 10.1007/s11033-014-3058-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 01/04/2014] [Indexed: 11/28/2022]
Abstract
Based on the nested case-control study cohort and gene expression profile, we have picked up a subset of six genes to distinguish the leukemia group and control group stably. ATG3 is the only down regulated gene. This research is to investigate the effect of ATG3 gene over expression by lentivirus on SKM-1 cell line and myelodysplastic syndrome to leukemic transformation. Human SKM-1 cells were transfected with ATG3-GFP recombinant lentiviral vectors and compared with cells transfected with GFP lentiviral vectors. Western blot was performed to detect the ATG3 protein. Cell proliferation was assessed by cell counting kit-8. Cell vitality was tested by Trypan Blue. Cell apoptosis was determined by Annexin V Apoptosis Detection Kit APC. Observe and compare the changes on growth curve, cell vitality and cell apoptosis. After 72 h of transfection, satisfactory transfection efficiency (> 90 %) was observed. SKM-1 cell line showed a statistically significant (P < 0.05) overexpression of ATG3, parallel to significantly (P < 0.05) inhibited cell proliferation. The cell vitality of ATG3 overexpression was significantly (P < 0.05) lower than negative control. Cell apoptosis analysis by flow cytometer demonstrated decreased proportion of early apoptosis and increased that of late apoptosis and death (P < 0.05). Over expressed ATG3 gene and protein, the SKM-1 cell line was inhibited in proliferation and cell vitality. It was promoted from early apoptosis to late apoptosis and death. The malignancy of SKM-1 cell line was decreased after transfection. ATG3 gene and its gene family may play an important role in transformation of myelodysplastic syndrome.
Collapse
Affiliation(s)
- Lin Wang
- Department of Hematology, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | | | | | | | | | | |
Collapse
|
50
|
Puleston DJ, Simon AK. Autophagy in the immune system. Immunology 2014; 141:1-8. [PMID: 23991647 PMCID: PMC3893844 DOI: 10.1111/imm.12165] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an intracellular homeostatic mechanism important for the degradation of waste components from the cytoplasm in acidic lysosomal compartments. Originally, surplus parts of the cytoplasm that acted as targets for autophagy were thought to comprise cellular organelles and proteins, but this has now extended to include a range of pathogens with particular emphasis on intracellular bacteria. The finding that autophagy can sequester intracellular bacteria and mediate their destruction has opened the door to a wider role for autophagy as an effector arm of the immune system. In innate immunity, autophagy works downstream of pattern recognition receptors where it facilitates a number of effector responses, including cytokine production and phagocytosis. Autophagy is also able to intersect pathways of innate and adaptive immunity through its potential to deliver antigens for antigen presentation. Autophagy provides a substantial source of antigens for loading onto MHC class II molecules and it may be important in dendritic cells for cross-priming to CD8(+) T cells. In lymphocytes, autophagy is essential for cell survival and homeostasis, particularly in T cells. In the thymus, autophagy can modulate the selection of certain CD4(+) T-cell clones while in the bone marrow autophagy is needed for B-cell development at specific stages. However, large holes exist in our knowledge as to how autophagy regulates, and is regulated by, the immune system and it is important to now apply what we have gleaned from in vitro studies to how autophagy operates in vivo in the setting of natural infection.
Collapse
Affiliation(s)
- Daniel J Puleston
- MRC Human Immunology Unit, The Weatherall Institute of Molecular Medicine, University of OxfordOxford, UK
| | - Anna Katharina Simon
- MRC Human Immunology Unit, The Weatherall Institute of Molecular Medicine, University of OxfordOxford, UK
- The BRC Translational Immunology Lab, NIHR, The John Radcliffe Hospital, University of OxfordOxford, UK
| |
Collapse
|