1
|
Wang HD, Lv CL, Feng L, Guo JX, Zhao SY, Jiang P. The role of autophagy in brain health and disease: Insights into exosome and autophagy interactions. Heliyon 2024; 10:e38959. [PMID: 39524893 PMCID: PMC11546156 DOI: 10.1016/j.heliyon.2024.e38959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2023] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Effective management of cellular components is essential for maintaining brain health, and studies have identified several crucial biological processes in the brain. Among these, autophagy and the role of exosomes in cellular communication are critical for brain health and disease. The interaction between autophagy and exosomes in the nervous system, as well as their contributions to brain damage, have garnered significant attention. This review summarizes that exosomes and their cargoes have been implicated in the autophagy process in the pathophysiology of nervous system diseases. Furthermore, the onset and progression of neurological disorders may be affected by autophagy regulation of the secretion and release of exosomes. These findings may provide new insights into the potential mechanism by which autophagy mediates different exosome secretion and release, as well as the valuable biomedical applications of exosomes in the prevention and treatment of various brain diseases by targeting autophagy.
Collapse
Affiliation(s)
- Hai-Dong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/Nanjing Medical University Kangda College First Affiliated Hospital/The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Chao-Liang Lv
- Department of Spine Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Jin-Xiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Shi-Yuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
2
|
Niharika, Garg M. Understanding the autophagic functions in cancer stem cell maintenance and therapy resistance. Expert Rev Mol Med 2024; 26:e23. [PMID: 39375840 PMCID: PMC11488345 DOI: 10.1017/erm.2024.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2023] [Revised: 12/25/2023] [Accepted: 06/25/2024] [Indexed: 10/09/2024]
Abstract
Complex tumour ecosystem comprising tumour cells and its associated tumour microenvironment (TME) constantly influence the tumoural behaviour and ultimately impact therapy failure, disease progression, recurrence and poor overall survival of patients. Crosstalk between tumour cells and TME amplifies the complexity by creating metabolic changes such as hypoxic environment and nutrient fluctuations. These changes in TME initiate stem cell-like programmes in cancer cells, contribute to tumoural heterogeneity and increase tumour robustness. Recent studies demonstrate the multifaceted role of autophagy in promoting fibroblast production, stemness, cancer cell survival during longer periods of dormancy, eventual growth of metastatic disease and disease resistance. Recent ongoing studies examine autophagy/mitophagy as a powerful survival strategy in response to environmental stress including nutrient deprivation, hypoxia and environmental stress in TME. It prevents irreversible senescence, promotes dormant stem-like state, induces epithelial-mesenchymal transition and increases migratory and invasive potential of tumour cells. The present review discusses various theories and mechanisms behind the autophagy-dependent induction of cancer stem cell (CSC) phenotype. Given the role of autophagic functions in CSC aggressiveness and therapeutic resistance, various mechanisms and studies based on suppressing cellular plasticity by blocking autophagy as a powerful therapeutic strategy to kill tumour cells are discussed.
Collapse
Affiliation(s)
- Niharika
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
3
|
Abstract
Microglia play key roles in the post-ischemic inflammatory response and damaged tissue removal reacting rapidly to the disturbances caused by ischemia and working to restore the lost homeostasis. However, the modified environment, encompassing ionic imbalances, disruption of crucial neuron-microglia interactions, spreading depolarization, and generation of danger signals from necrotic neurons, induce morphological and phenotypic shifts in microglia. This leads them to adopt a proinflammatory profile and heighten their phagocytic activity. From day three post-ischemia, macrophages infiltrate the necrotic core while microglia amass at the periphery. Further, inflammation prompts a metabolic shift favoring glycolysis, the pentose-phosphate shunt, and lipid synthesis. These shifts, combined with phagocytic lipid intake, drive lipid droplet biogenesis, fuel anabolism, and enable microglia proliferation. Proliferating microglia release trophic factors contributing to protection and repair. However, some microglia accumulate lipids persistently and transform into dysfunctional and potentially harmful foam cells. Studies also showed microglia that either display impaired apoptotic cell clearance, or eliminate synapses, viable neurons, or endothelial cells. Yet, it will be essential to elucidate the viability of engulfed cells, the features of the local environment, the extent of tissue damage, and the temporal sequence. Ischemia provides a rich variety of region- and injury-dependent stimuli for microglia, evolving with time and generating distinct microglia phenotypes including those exhibiting proinflammatory or dysfunctional traits and others showing pro-repair features. Accurate profiling of microglia phenotypes, alongside with a more precise understanding of the associated post-ischemic tissue conditions, is a necessary step to serve as the potential foundation for focused interventions in human stroke.
Collapse
Affiliation(s)
- Anna M Planas
- Cerebrovascular Research Laboratory, Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Cerebrovascular Diseases, Area of Clinical and Experimental Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Barcelona, Spain
| |
Collapse
|
4
|
Fila M, Pawlowska E, Szczepanska J, Blasiak J. Autophagy may protect the brain against prolonged consequences of headache attacks: A narrative/hypothesis review. Headache 2023; 63:1154-1166. [PMID: 37638395 DOI: 10.1111/head.14625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/07/2023] [Revised: 06/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE To assess the potential of autophagy in migraine pathogenesis. BACKGROUND The interplay between neurons and microglial cells is important in migraine pathogenesis. Migraine-related effects, such as cortical spreading depolarization and release of calcitonin gene-related peptide, may initiate adenosine triphosphate (ATP)-mediating pro-nociceptive signaling in the meninges causing headaches. Such signaling may be induced by the interaction of ATP with purinergic receptor P2X 7 (P2X7R) on microglial cells leading to a Ca2+ -mediated pH increase in lysosomes and release of autolysosome-like vehicles from microglial cells indicating autophagy impairment. METHODS A search in PubMed was conducted with the use of the terms "migraine," "autophagy," "microglia," and "degradation" in different combinations. RESULTS Impaired autophagy in microglia may activate secretory autophagy and release of specific proteins, including brain-derived neurotrophic factor (BDNF), which can be also released through the pores induced by P2X7R activation in microglial cells. BDNF may be likewise released from microglial cells upon ATP- and Ca2+ -mediated activation of another purinergic receptor, P2X4R. BDNF released from microglia might induce autophagy in neurons to clear cellular debris produced by oxidative stress, which is induced in the brain as the response to migraine-related energy deficit. Therefore, migraine-related signaling may impair degradative autophagy, stimulate secretory autophagy in microglia, and degradative autophagy in neurons. These effects are mediated by purinergic receptors P2X4R and P2X7R, BDNF, ATP, and Ca2+ . CONCLUSION Different effects of migraine-related events on degradative autophagy in microglia and neurons may prevent prolonged changes in the brain related to headache attacks.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, Lodz, Poland
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, Lodz, Poland
| | - Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| |
Collapse
|
5
|
Huangfu H, Du S, Zhang H, Wang H, Zhang Y, Yang Z, Zhang X, Ren S, Chen S, Wang C, Zhang Y, Zhou Y. Facile engineering of resveratrol nanoparticles loaded with 20(S)-protopanaxadiol for the treatment of periodontitis by regulating the macrophage phenotype. NANOSCALE 2023; 15:7894-7908. [PMID: 37060139 DOI: 10.1039/d2nr06452a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/05/2023]
Abstract
Periodontitis is an inflammatory disease, mainly caused by the formation of a subgingival plaque biofilm. In recent years, growing attention has been paid to immunotherapy in the treatment of periodontitis, and the importance of communal intervention associated with macrophage polarization was emphasized. Herein, resveratrol (RES) and 20(S)-protopanaxadiol (PPD) were successfully self-assembled into RES@PPD nanoparticles (NPs) by the phenolic resin reaction. RES@PPD NPs have good stability and biocompatibility. The combined application of PPD and RES enhances the anti-inflammatory and antioxidant properties of nanocomposites, remarkably reduces the level of reactive oxygen species, and finally realizes the coordinated regulation of host immunity in periodontitis. The detailed mechanism is as follows: RES@PPD NPs inhibit M1 polarization of macrophages, promote M2 polarization by scavenging ROS, and then inhibit the NF-κB signalling pathway to regulate host immunity. In the animal model of periodontitis, RES@PPD NPs can remarkably decrease the level of pro-inflammatory cytokines, up-regulate the anti-inflammatory cytokines, and exhibit a profound therapeutic effect on local inflammation. Therefore, RES@PPD NPs are effective in antioxidation and anti-inflammation, thus providing a promising candidate drug for the treatment of periodontitis.
Collapse
Affiliation(s)
- Huimin Huangfu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Shulin Du
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Hao Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Hanchi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Zhen Yang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Xinwei Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Sicong Ren
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Siyu Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences Jilin University, Changchun, 130021, China.
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
6
|
Swamy K. Vascular normalization and immunotherapy: Spawning a virtuous cycle. Front Oncol 2022; 12:1002957. [PMID: 36276103 PMCID: PMC9582256 DOI: 10.3389/fonc.2022.1002957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Anti-angiogenics, radiotherapy (especially stereotactic body radiotherapy, SBRT)/chemotherapy, and immunotherapy form a critical trimodal approach in modern cancer therapy. The normalization window, however short, is the beachhead for the strategic initiation of a decipherable disruption of cancer cells. This opening can be the opportunity for designing controlled stepwise cancer cell death (CCD) and immunological augmentation. The next step is to induce immunogenic cell death (ICD) through chemotherapy/radiotherapy concurrently with the facilitation of professional phagocytosis. Immunotherapy at this stage, when interstitial pressure decreases considerably, leads to the improved perfusion of oxygen with solutes and improved immune-friendly pH and is additionally expected to open up the tumor microenvironment (TME) for a “flood” of tumor-infiltrating lymphocytes. Furthermore, there would be enhanced interaction in “hot” nodules and the incorporation of immune reaction in “cold” nodules. Simultaneously, the added adjuvant-assisted neoantigen–immune cell interaction will likely set in a virtuous cycle of CCD induction followed by tumor cell-specific antigenic reaction boosting CCD, in turn promoting the normalization of the vasculature, completing the loop. There should be a conscious concern to protect the extracellular matrix (ECM), which will nurture the long-term immunological cross-talk to discourage dormancy, which is as essential as obtaining a complete response in imaging. The caveat is that the available therapies should be appropriately ranked during the start of the treatment since the initial administration is the most opportune period. A fast-paced development in the nanomedicine field is likely to assist in all the steps enumerated.
Collapse
|
7
|
Zhang T, Liu Q, Gao W, Sehgal SA, Wu H. The multifaceted regulation of mitophagy by endogenous metabolites. Autophagy 2022; 18:1216-1239. [PMID: 34583624 PMCID: PMC9225590 DOI: 10.1080/15548627.2021.1975914] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/23/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/30/2022] Open
Abstract
Owing to the dominant functions of mitochondria in multiple cellular metabolisms and distinct types of regulated cell death, maintaining a functional mitochondrial network is fundamental for the cellular homeostasis and body fitness in response to physiological adaptations and stressed conditions. The process of mitophagy, in which the dysfunctional or superfluous mitochondria are selectively engulfed by autophagosome and subsequently degraded in lysosome, has been well formulated as one of the major mechanisms for mitochondrial quality control. To date, the PINK1-PRKN-dependent and receptors (including proteins and lipids)-dependent pathways have been characterized to determine the mitophagy in mammalian cells. The mitophagy is highly responsive to the dynamics of endogenous metabolites, including iron-, calcium-, glycolysis-TCA-, NAD+-, amino acids-, fatty acids-, and cAMP-associated metabolites. Herein, we summarize the recent advances toward the molecular details of mitophagy regulation in mammalian cells. We also highlight the key regulations of mammalian mitophagy by endogenous metabolites, shed new light on the bidirectional interplay between mitophagy and cellular metabolisms, with attempting to provide a perspective insight into the nutritional intervention of metabolic disorders with mitophagy deficit.Abbreviations: acetyl-CoA: acetyl-coenzyme A; ACO1: aconitase 1; ADCYs: adenylate cyclases; AMPK: AMP-activated protein kinase; ATM: ATM serine/threonine kinase; BCL2L1: BCL2 like 1; BCL2L13: BCL2 like 13; BNIP3: BCL2 interacting protein 3; BNIP3L: BCL2 interacting protein 3 like; Ca2+: calcium ion; CALCOCO2: calcium binding and coiled-coil domain 2; CANX: calnexin; CO: carbon monoxide; CYCS: cytochrome c, somatic; DFP: deferiprone; DNM1L: dynamin 1 like; ER: endoplasmic reticulum; FKBP8: FKBP prolyl isomerase 8; FOXO3: forkhead box O3; FTMT: ferritin mitochondrial; FUNDC1: FUN14 domain containing 1; GABA: γ-aminobutyric acid; GSH: glutathione; HIF1A: hypoxia inducible factor 1 subunit alpha; IMMT: inner membrane mitochondrial protein; IRP1: iron regulatory protein 1; ISC: iron-sulfur cluster; ITPR2: inositol 1,4,5-trisphosphate type 2 receptor; KMO: kynurenine 3-monooxygenase; LIR: LC3 interacting region; MAM: mitochondria-associated membrane; MAP1LC3: microtubule associated protein 1 light chain 3; MFNs: mitofusins; mitophagy: mitochondrial autophagy; mPTP: mitochondrial permeability transition pore; MTOR: mechanistic target of rapamycin kinase; NAD+: nicotinamide adenine dinucleotide; NAM: nicotinamide; NMN: nicotinamide mononucleotide; NO: nitric oxide; NPA: Niemann-Pick type A; NR: nicotinamide riboside; NR4A1: nuclear receptor subfamily 4 group A member 1; NRF1: nuclear respiratory factor 1; OPA1: OPA1 mitochondrial dynamin like GTPase; OPTN: optineurin; PARL: presenilin associated rhomboid like; PARPs: poly(ADP-ribose) polymerases; PC: phosphatidylcholine; PHB2: prohibitin 2; PINK1: PTEN induced kinase 1; PPARG: peroxisome proliferator activated receptor gamma; PPARGC1A: PPARG coactivator 1 alpha; PRKA: protein kinase AMP-activated; PRKDC: protein kinase, DNA-activated, catalytic subunit; PRKN: parkin RBR E3 ubiquitin protein ligase; RHOT: ras homolog family member T; ROS: reactive oxygen species; SIRTs: sirtuins; STK11: serine/threonine kinase 11; TCA: tricarboxylic acid; TP53: tumor protein p53; ULK1: unc-51 like autophagy activating kinase 1; VDAC1: voltage dependent anion channel 1.
Collapse
Affiliation(s)
- Ting Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, China
| | - Qian Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, China
| | - Weihua Gao
- Hubei Hongshan Laboratory, Wuhan, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | | | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
8
|
Hew JJ, Parungao RJ, Mooney CP, Smyth JK, Kim S, Tsai KHY, Shi H, Chong C, Chan RCF, Attia B, Nicholls C, Li Z, Solon-Biet SM, Le Couteur DG, Simpson SJ, Jeschke MG, Maitz PK, Wang Y. Low-protein diet accelerates wound healing in mice post-acute injury. BURNS & TRAUMA 2021; 9:tkab010. [PMID: 34377708 PMCID: PMC8350350 DOI: 10.1093/burnst/tkab010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/08/2020] [Revised: 12/06/2020] [Accepted: 03/11/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND Wound healing processes are influenced by macronutrient intake (protein, carbohydrate and fat). The most favourable diet for cutaneous wound healing is not known, although high-protein diets are currently favoured clinically. This experimental study investigates the optimal macronutrient balance for cutaneous wound healing using a mouse model and the Geometric Framework, a nutrient modelling method, capable of analyzing the individual and interactive effects of a wide spectrum of macronutrient intake. METHODS Two adjacent and identical full-thickness skin excisions (1 cm2) were surgically created on the dorsal area of male C57BL/6 mice. Mice were then allocated to one of 12 high-energy diets that varied in protein, carbohydrate and fat content. In select diets, wound healing processes, cytokine expression, energy expenditure, body composition, muscle and fat reserves were assessed. RESULTS Using the Geometric Framework, we show that a low-protein intake, coupled with a balanced intake of carbohydrate and fat is optimal for wound healing. Mice fed a low-protein diet progressed quickly through wound healing stages with favourable wound inflammatory cytokine expression and significantly accelerated collagen production. These local processes were associated with an increased early systemic inflammatory response and a higher overall energy expenditure, related to metabolic changes occurring in key macronutrient reserves in lean body mass and fat depots. CONCLUSIONS The results suggest that a low-protein diet may have a greater potential to accelerate wound healing than the current clinically used high-protein diets.
Collapse
Affiliation(s)
- Jonathan J Hew
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Roxanne J Parungao
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Craig P Mooney
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Julian K Smyth
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Sarah Kim
- Bone Biology Group, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Kevin H-Y Tsai
- Adrenal Steroids Laboratory, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Huaikai Shi
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Cassandra Chong
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Renee C F Chan
- Electron Microscopy Unit, Anatomical Pathology, Concord Hospital, Sydney, Australia 2139
| | - Beba Attia
- Electron Microscopy Unit, Anatomical Pathology, Concord Hospital, Sydney, Australia 2139
| | - Caroline Nicholls
- Burns Unit, Concord Repatriation General Hospital, Concord, Australia 2139
| | - Zhe Li
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
- Burns Unit, Concord Repatriation General Hospital, Concord, Australia 2139
| | - Samantha M Solon-Biet
- Ageing and Alzheimer Institute and ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Australia 2006
| | - David G Le Couteur
- Ageing and Alzheimer Institute and ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
| | - Stephen J Simpson
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Australia 2006
| | - Marc G Jeschke
- Sunnybrook Research Institute, Toronto, Ontario, Canada, M4N 3M5
| | - Peter K Maitz
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
- Burns Unit, Concord Repatriation General Hospital, Concord, Australia 2139
| | - Yiwei Wang
- Burns Research and Reconstructive Surgery, ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney, Australia 2139
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, PR China 210023
| |
Collapse
|
9
|
Chen C, Gao H, Su X. Autophagy-related signaling pathways are involved in cancer (Review). Exp Ther Med 2021; 22:710. [PMID: 34007319 PMCID: PMC8120650 DOI: 10.3892/etm.2021.10142] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/11/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a self-digestion process in cells that can maintain energy homeostasis under normal circumstances. However, misfolded proteins, damaged mitochondria and other unwanted components in cells can be decomposed and reused via autophagy in some specific cases (including hypoxic stress, low energy states or nutrient deprivation). Therefore, autophagy serves a positive role in cell survival and growth. However, excessive autophagy may lead to apoptosis. Furthermore, abnormal autophagy may lead to carcinogenesis and promote tumorigenesis in normal cells. In tumor cells, autophagy may provide the energy required for excessive proliferation, promote the growth of cancer cells, and evade apoptosis caused by certain treatments, including radiotherapy and chemotherapy, resulting in increased treatment resistance and drug resistance. On the other hand, autophagy leads to an insufficient nutrient supply in cancer cells and the destruction of energy homeostasis, thereby inducing cancer cell apoptosis. Therefore, understanding the mechanism of the double-edged sword of autophagy is crucial for the treatment of cancer. The present review summarizes the signaling pathways and key factors involved in autophagy and cancer to provide possible strategies for treating tumors.
Collapse
Affiliation(s)
- Caixia Chen
- Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Hui Gao
- Department of Thoracic Surgery, Inner Mongolia Autonomous Region Cancer Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xiulan Su
- Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| |
Collapse
|
10
|
Minasyan H. Oxycytosis and the role of triboelectricity and oxidation in bacteria clearing from the bloodstream. Eur J Microbiol Immunol (Bp) 2021; 11:23-28. [PMID: 34019486 PMCID: PMC8287977 DOI: 10.1556/1886.2021.00008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/07/2021] [Accepted: 04/25/2021] [Indexed: 11/19/2022] Open
Abstract
Until recently, little was known about the mechanism for killing and clearing bacteria from the bloodstream. Leukocyte phagocytosis could not be a mechanism for catching, killing and removing bacteria from the bloodstream because of many reasons. Recently accumulated data have led to the conclusion that in bacteremia, bacteria are quickly removed from the blood and erythrocytes are the main cells that capture, kill and remove bacteria. Data were also obtained that erythrocytes catch bacteria by triboelectric charge attraction and kill them by oxygen released from oxyhemoglobin. This phenomenon has been named oxycytosis by analogy with the term phagocytosis. Oxycytosis has been discussed in a number of published articles, but the specific mechanism of triboelectric charging and the mechanism of killing bacteria by oxidation, have not yet been detailed. The purpose of this review is to provide a more detailed explanation of the process of triboelectric charging and capture of bacteria by erythrocytes and destruction of bacteria by oxidation. For the first time, the review presents various variants of oxycytosis (two-stage, three-stage, multi-stage), depending on the resistance of the pathogen to oxidation. The review also discusses the biological significance of oxycytosis and its impact on the understanding of bacteremia and sepsis.
Collapse
|
11
|
Aoto K, Kato M, Akita T, Nakashima M, Mutoh H, Akasaka N, Tohyama J, Nomura Y, Hoshino K, Ago Y, Tanaka R, Epstein O, Ben-Haim R, Heyman E, Miyazaki T, Belal H, Takabayashi S, Ohba C, Takata A, Mizuguchi T, Miyatake S, Miyake N, Fukuda A, Matsumoto N, Saitsu H. ATP6V0A1 encoding the a1-subunit of the V0 domain of vacuolar H +-ATPases is essential for brain development in humans and mice. Nat Commun 2021; 12:2107. [PMID: 33833240 PMCID: PMC8032687 DOI: 10.1038/s41467-021-22389-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/10/2019] [Accepted: 03/05/2021] [Indexed: 02/01/2023] Open
Abstract
Vacuolar H+-ATPases (V-ATPases) transport protons across cellular membranes to acidify various organelles. ATP6V0A1 encodes the a1-subunit of the V0 domain of V-ATPases, which is strongly expressed in neurons. However, its role in brain development is unknown. Here we report four individuals with developmental and epileptic encephalopathy with ATP6V0A1 variants: two individuals with a de novo missense variant (R741Q) and the other two individuals with biallelic variants comprising one almost complete loss-of-function variant and one missense variant (A512P and N534D). Lysosomal acidification is significantly impaired in cell lines expressing three missense ATP6V0A1 mutants. Homozygous mutant mice harboring human R741Q (Atp6v0a1R741Q) and A512P (Atp6v0a1A512P) variants show embryonic lethality and early postnatal mortality, respectively, suggesting that R741Q affects V-ATPase function more severely. Lysosomal dysfunction resulting in cell death, accumulated autophagosomes and lysosomes, reduced mTORC1 signaling and synaptic connectivity, and lowered neurotransmitter contents of synaptic vesicles are observed in the brains of Atp6v0a1A512P/A512P mice. These findings demonstrate the essential roles of ATP6V0A1/Atp6v0a1 in neuronal development in terms of integrity and connectivity of neurons in both humans and mice. A member of the vacuolar H+-ATPase family, ATP6V0A1 is involved in lysosomal activity. Here, the authors report that ATP6V0A1 variants identified in individuals with developmental and epileptic encephalopathy are associated with impairment of lysosomal acidification, autophagy and mTORC1 signaling, suggesting an essential role of ATP6V0A1 in brain development.
Collapse
Affiliation(s)
- Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Tenpei Akita
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroki Mutoh
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Akasaka
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, Japan.,Department of Pediatrics, Niigata Prefecture Hamagumi Medical Rehabilitation Center for Disabled Children, Niigata, Japan
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, Japan
| | - Yoshiko Nomura
- Segawa Neurological Clinic for Children, Tokyo, Japan.,Yoshiko Nomura Neurological Clinic for Children, Tokyo, Japan
| | - Kyoko Hoshino
- Segawa Neurological Clinic for Children, Tokyo, Japan.,Segawa Memorial Neurological Clinic for Children, Tokyo, Japan
| | - Yasuhiko Ago
- Department of Neonatology, Ibaraki Children's Hospital, Mito, Japan.,Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Ryuta Tanaka
- Ibaraki Pediatric Education and Training Station, University of Tsukuba, Mito, Japan
| | - Orna Epstein
- Pediatric Neurology and Development Center, Shamir Medical Center, Tzrifin, Beer Yaakov, Israel
| | - Revital Ben-Haim
- Pediatric Neurology and Development Center, Shamir Medical Center, Tzrifin, Beer Yaakov, Israel
| | - Eli Heyman
- Pediatric Neurology and Development Center, Shamir Medical Center, Tzrifin, Beer Yaakov, Israel
| | - Takehiro Miyazaki
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hazrat Belal
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shuji Takabayashi
- Laboratory Animal Facilities & Services, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chihiro Ohba
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
12
|
Human Umbilical Cord: Information Mine in Sex-Specific Medicine. Life (Basel) 2021; 11:life11010052. [PMID: 33451112 PMCID: PMC7828611 DOI: 10.3390/life11010052] [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] [Academic Contribution Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Biological differences between sexes should be considered in all stages of research, as sexual dimorphism starts in utero leading to sex-specific fetal programming. In numerous biomedical fields, there is still a lack of stratification by sex despite primary cultured cells retaining memory of the sex and of the donor. The sex of donors in biological research must be known because variations in cells and cellular components can be used as endpoints, biomarkers and/or targets of pharmacological studies. This selective review focuses on the current findings regarding sex differences observed in the umbilical cord, a widely used source of research samples, both in the blood and in the circulating cells, as well as in the different cellular models obtainable from it. Moreover, an overview on sex differences in fetal programming is reported. As it emerges that the sex variable is still often forgotten in experimental models, we suggest that it should be mandatory to adopt sex-oriented research, because only awareness of these issues can lead to innovative research.
Collapse
|
13
|
Jabir MS, Hussien AA, Sulaiman GM, Yaseen NY, Dewir YH, Alwahibi MS, Soliman DA, Rizwana H. Green synthesis of silver nanoparticles from Eriobotrya japonica extract: a promising approach against cancer cells proliferation, inflammation, allergic disorders and phagocytosis induction. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 49:48-60. [DOI: 10.1080/21691401.2020.1867152] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Affiliation(s)
- Majid S. Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Aya A. Hussien
- College of Dentistry, Mustansiriyah University, Baghdad, Iraq
| | | | - Nahi Y. Yaseen
- Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq
| | - Yaser H. Dewir
- Plant Production Department, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
- Faculty of Agriculture, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Mona S. Alwahibi
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Dina A. Soliman
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Humaira Rizwana
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Hu Y, Zhang J, Liu Q, Ke M, Li J, Suo W, Guo W, Ma A. Torin2 inhibits the EGFR-TKI resistant Non-Small Lung Cancer cell proliferation through negative feedback regulation of Akt/mTOR signaling. J Cancer 2020; 11:5746-5757. [PMID: 32913468 PMCID: PMC7477446 DOI: 10.7150/jca.37417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/08/2019] [Accepted: 07/17/2020] [Indexed: 01/06/2023] Open
Abstract
It is known that mammalian target of rapamycin (mTOR) signaling plays an important role in NSCLC cells proliferation. Torin2 is a second-generation ATP-competitive inhibitor which is selective for mTOR activity. In this study, we investigated whether torin2 was effective against lung cancer cells, especially EGFR-TKIs resistant NSCLC cells. We found that torin2 dramatically inhibited EGFR-TKI resistant cells viability in vitro. In xenograft model, torin2 treatment significantly reduced the volume and weight of xenograft tumor in the erlotinib resistant PC9/E cells. Additionally, autophagy protein of phosphatidylethanolamine-modified microtubule-associated protein light-chain 3II/I (LC3II/I) increased in PC9/E after torin2 treatment. Torin2 blocked the level of phosphorylated S6 and the phosphorylation of Akt at both T308 and S473 sites compared with erlotinib treatment. Furthermore, TUNEL assay showed that apoptosis of tumor tissue increased significantly in the torin2 treatment group. Immunohistochemical analysis demonstrated that tumor angiogenesis was obviously inhibited by torin2 treatment in EGFR-TKI resistant group. Collectively, our results suggested that torin2 could inhibit the NSCLC cells proliferation by negative feedback regulation of Akt/mTOR signaling and inducing autophagy. This suggests that torin2 could be a novel therapeutic approach for EGFR-TKI resistant NSCLC.
Collapse
Affiliation(s)
- Yi Hu
- Department of Clinical Laboratory, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Ji Zhang
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, China
| | - Qun Liu
- Department of Respiratory and Critical Medicine, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Mingyao Ke
- Department of Respiratory and Critical Medicine, The secondary hospital of Xiamen Medicine school, Xiamen, China
| | - Jiurong Li
- Department of Respiratory and Critical Medicine, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Wenhao Suo
- Department of Pathology, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Weixi Guo
- Department of Thoracic Surgery, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Aiping Ma
- Department of Respiratory and Critical Medicine, The first affiliated hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
15
|
Bae HR, Leung PSC, Hodge DL, Fenimore JM, Jeon SM, Thovarai V, Dzutsev A, Welcher AA, Boedigheimer M, Damore MA, Choi MS, Fravell RA, Trinchieri G, Gershwin ME, Young HA. Multi-omics: Differential expression of IFN-γ results in distinctive mechanistic features linking chronic inflammation, gut dysbiosis, and autoimmune diseases. J Autoimmun 2020; 111:102436. [PMID: 32220507 PMCID: PMC7266723 DOI: 10.1016/j.jaut.2020.102436] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2019] [Revised: 02/26/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
Low grade, chronic inflammation is a critical risk factor for immunologic dysfunction including autoimmune diseases. However, the multiplicity of complex mechanisms and lack of relevant murine models limit our understanding of the precise role of chronic inflammation. To address these hurdles, we took advantage of multi-omics data and a unique murine model with a low but chronic expression of IFN-γ, generated by replacement of the AU-rich element (ARE) in the 3' UTR region of IFN-γ mRNA with random nucleotides. Herein, we demonstrate that low but differential expression of IFN-γ in mice by homozygous or heterozygous ARE replacement triggers distinctive gut microbial alterations, of which alteration is female-biased with autoimmune-associated microbiota. Metabolomics data indicates that gut microbiota-dependent metabolites have more robust sex-differences than microbiome profiling, particularly those involved in fatty acid oxidation and nuclear receptor signaling. More importantly, homozygous ARE-Del mice have dramatic changes in tryptophan metabolism, bile acid and long-chain lipid metabolism, which interact with gut microbiota and nuclear receptor signaling similarly with sex-dependent metabolites. Consistent with these findings, nuclear receptor signaling, encompassing molecules such as PPARs, FXR, and LXRs, was detectable as a top canonical pathway in comparison of blood and tissue-specific gene expression between female homozygous vs heterozygous ARE-Del mice. Further analysis implies that dysregulated autophagy in macrophages is critical for breaking self-tolerance and gut homeostasis, while pathways interact with nuclear receptor signaling to regulate inflammatory responses. Overall, pathway-based integration of multi-omics data provides systemic and cellular insights about how chronic inflammation driven by IFN-γ results in the development of autoimmune diseases with specific etiopathological features.
Collapse
Affiliation(s)
- Heekyong R Bae
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA.
| | - Deborah L Hodge
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - John M Fenimore
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Seon-Min Jeon
- Center for Food and Nutritional Genomics Research, Department of Food Science and Nutrition, Kyungpook National University, Daegu, Republic of Korea.
| | - Vishal Thovarai
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Amiran Dzutsev
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | | | | | | | - Myung-Sook Choi
- Center for Food and Nutritional Genomics Research, Department of Food Science and Nutrition, Kyungpook National University, Daegu, Republic of Korea.
| | - Richard A Fravell
- Department of Immunology, Yale School of Medicine, New Haven, CT, USA.
| | - Giorgio Trinchieri
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA.
| |
Collapse
|
16
|
Deng M, Huang L, Zhong X. β‑asarone modulates Beclin‑1, LC3 and p62 expression to attenuate Aβ40 and Aβ42 levels in APP/PS1 transgenic mice with Alzheimer's disease. Mol Med Rep 2020; 21:2095-2102. [PMID: 32186763 PMCID: PMC7115210 DOI: 10.3892/mmr.2020.11026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/09/2018] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease in the elderly population. Autophagy is a well-known regulator of neurodegenerative diseases and β-asarone has been discovered to have certain neuropharmacological effects. Thus, the present study aimed to analyze the potential effects of β-asarone in AD and its possible mechanism of action in relation to autophagy. The present study investigated the effects of β-asarone on the number of senile plaques and amyloid β(Aβ)40, Aβ42, amyloid precursor protein (APP) and Beclin-1 mRNA levels in the hippocampus of APP/presenilin-1 (PS1) transgenic mice. The possible mechanism of β-asarone on autophagy-related proteins, including Beclin-1, light chain (LC)3A, LC3B and p62 levels, and the number of autophagosomes was also investigated. Mice were divided into a normal control group, a model group, a β-asarone-treated group, a 3-MA-treated group and a rapamycin-treated group. Treatments were continuously administered to all mice for 30 days by intragastric administration. The mice, including those in the normal and model control groups, were given equal volumes of saline. It was demonstrated that β-asarone treatment reduced the number of senile plaques and autophagosomes, and decreased Aβ40, Aβ42, APP and Beclin-1 expression in the hippocampus of model mice compared with untreated model mice. β-asarone also inhibited LC3A/B expression levels, but increased p62 expression. It was deduced that the neuroprotective effects of β-asarone in APP/PS1 transgenic mice resulted from its inhibition of autophagy. In conclusion, the data suggested that β-asarone should be explored further as a potential therapeutic agent in AD.
Collapse
Affiliation(s)
- Minzhen Deng
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Liping Huang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang, Guangdong 524048, P.R. China
| | - Xiaoqin Zhong
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
17
|
Melcarne C, Ramond E, Dudzic J, Bretscher AJ, Kurucz É, Andó I, Lemaitre B. Two Nimrod receptors, NimC1 and Eater, synergistically contribute to bacterial phagocytosis in Drosophila melanogaster. FEBS J 2019; 286:2670-2691. [PMID: 30993828 PMCID: PMC6852320 DOI: 10.1111/febs.14857] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2018] [Revised: 03/19/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022]
Abstract
Eater and NimC1 are transmembrane receptors of the Drosophila Nimrod family, specifically expressed in haemocytes, the insect blood cells. Previous ex vivo and in vivoRNAi studies have pointed to their role in the phagocytosis of bacteria. Here, we have created a novel NimC1 null mutant to re-evaluate the role of NimC1, alone or in combination with Eater, in the cellular immune response. We show that NimC1 functions as an adhesion molecule ex vivo, but in contrast to Eater it is not required for haemocyte sessility in vivo. Ex vivo phagocytosis assays and electron microscopy experiments confirmed that Eater is the main phagocytic receptor for Gram-positive, but not Gram-negative bacteria, and contributes to microbe tethering to haemocytes. Surprisingly, NimC1 deletion did not impair phagocytosis of bacteria, nor their adhesion to the haemocytes. However, phagocytosis of both types of bacteria was almost abolished in NimC11 ;eater1 haemocytes. This indicates that both receptors contribute synergistically to the phagocytosis of bacteria, but that Eater can bypass the requirement for NimC1. Finally, we uncovered that NimC1, but not Eater, is essential for uptake of latex beads and zymosan particles. We conclude that Eater and NimC1 are the two main receptors for phagocytosis of bacteria in Drosophila, and that each receptor likely plays distinct roles in microbial uptake.
Collapse
Affiliation(s)
- Claudia Melcarne
- Global Health InstituteSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)Switzerland
| | - Elodie Ramond
- Global Health InstituteSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)Switzerland
| | - Jan Dudzic
- Global Health InstituteSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)Switzerland
| | - Andrew J. Bretscher
- Global Health InstituteSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)Switzerland
| | - Éva Kurucz
- Institute of GeneticsBiological Research Centre of the Hungarian Academy of SciencesSzegedHungary
| | - István Andó
- Institute of GeneticsBiological Research Centre of the Hungarian Academy of SciencesSzegedHungary
| | - Bruno Lemaitre
- Global Health InstituteSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)Switzerland
| |
Collapse
|
18
|
Oh DS, Lee HK. Autophagy protein ATG5 regulates CD36 expression and anti-tumor MHC class II antigen presentation in dendritic cells. Autophagy 2019; 15:2091-2106. [PMID: 30900506 DOI: 10.1080/15548627.2019.1596493] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022] Open
Abstract
Macroautophagy/autophagy has been implicated in cytoplasmic and viral antigen presentation on major histocompatibility complex (MHC) class II molecules. However, the role of autophagy in the presentation of phagocytized tumor-associated antigens in vivo remains unclear. Following the administration of apoptotic tumor cells and in vivo chemotherapy, mice with a dendritic cell-specific deletion of Atg5, a key autophagy gene, exhibit reduced CD4+ T-cell priming but not CD8+ cytotoxic T-cell priming. Interestingly, Atg5-deficient dendritic cells have an elevated expression of scavenger receptor CD36 and show excessive lipid accumulation. Atg5-deficient dendritic cells increased CD36-dependent phagocytosis of apoptotic tumor cells. CD36 blockade ameliorates elevated phagocytosis and increases CD4+ T-cell priming in dendritic cells; intratumoral CD36 blockade inhibits tumor growth. Our results demonstrate that Atg5 is required for proper antigen phagocytosis and presentation to MHC class II via modulation of CD36 in dendritic cells and may be a future therapeutic target for anti-tumor therapy.Abbreviations: APC: antigen-presenting cell; ATG: autophagy-related; BMDC: bone marrow-derived dendritic cell; BODIPY: 4,4-difluoro-1,3,5,7,8-pentamethyl-4-bora-3a,4a-diaza-s-indacene; CSFE: carboxyfluorescein diacetate succinimidyl ester; DAPI: 4',6-diamidino-2-phenylindole; IFNG/IFN-γ: interferon gamma; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MHC: major histocompatibility complex; NLDC: neonatal liver-derived dendritic cell; PDCD1/PD-1: programmed cell death 1; PI: propidium iodide; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; SERPINB/OVA: serine (or cysteine) peptidase inhibitor, clade B; TIMD4/TIM-4: T cell immunoglobulin and mucin domain containing 4.
Collapse
Affiliation(s)
- Dong Sun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, Republic of Korea.,KAIST Institute for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
19
|
Zhang HL, Zhu YM, Zhou XY. Coordination of Autophagy and Other Cellular Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:697-727. [PMID: 31777007 DOI: 10.1007/978-981-15-0602-4_30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
Conventionally, autophagy (=self-eating) is thought to be a catabolic cellular process that is responsible for regulating cell homeostasis. However, the newly evidence have expanded the range of the impact of autophagy in biology. Autophagy interplays with endocytosis through shared factors such as phosphatidylinositol 3 kinase complex (PI(3)K complex), autophagy associated gene (Atg), and lysosome. Autophagy and phagocytosis orchestrate in maintaining homeostasis, in MHC class II antigen processing, in the removal of pathogens, in cell death, immunity, and inflammation. There are numerous cross talks of autophagy with biosynthetic processes such as conventional and unconventional secretion of biologically active cargo and trafficking of integral membrane proteins, as well as the exosome secretion. There are also links between autophagy and trafficking events from plasma membrane, including lateral plasma membrane proteins connexins, cell connections, and ciliogenesis.
Collapse
Affiliation(s)
- Hui-Ling Zhang
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China.
| | - Yong-Ming Zhu
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China
| | - Xian-Yong Zhou
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
20
|
Jahagirdar PS, Gupta PK, Kulkarni SP, Devarajan PV. Polymeric curcumin nanoparticles by a facile in situ method for macrophage targeted delivery. Bioeng Transl Med 2019; 4:141-151. [PMID: 30680325 PMCID: PMC6336664 DOI: 10.1002/btm2.10112] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 01/05/2023] Open
Abstract
Targeting macrophages is a promising strategy for improved therapy of intracellular infections as macrophages exhibit rapid phagocytosis of particles >200 nm. Entrapment of Curcumin (CUR) in nanocarriers could provide bioenhancement and macrophage targeting. We present a simple and facile in situ nanoprecipitation approach for instantaneous and on-site generation of curcumin nanoparticles (ISCurNP). ISCurNP optimised by Box-Behnken design exhibited average size of 208.25 ± 7.55 nm and entrapment efficiency of 90.16 ± 1.17%. Differential scanning calorimetry and X-Ray diffraction confirmed amorphization of CUR in ISCurNP. Sustained release was observed over 72 hr in vitro at lysosomal pH 4.5. Rapid and high uptake in RAW 264.7 macrophages was confirmed by flow cytometry and high performance liquid chromatography. Confocal microscopy established localisation of ISCurNP in lysosomal compartment. The facile in situ nanoprecipitation method provides simple, scalable technology to enable macrophage targeted delivery of CUR, with great promise for improved therapy of intracellular infections.
Collapse
Affiliation(s)
- Priyanka S. Jahagirdar
- Dept. of Pharmaceutical Sciences and TechnologyInstitute of Chemical TechnologyMatungaMumbai, MHIndia
| | - Pramod K. Gupta
- Radiation Medicine CentreBhabha Atomic Research CentreParelMumbai, MHIndia
| | - Savita P. Kulkarni
- Radiation Medicine CentreBhabha Atomic Research CentreParelMumbai, MHIndia
| | - Padma V. Devarajan
- Dept. of Pharmaceutical Sciences and TechnologyInstitute of Chemical TechnologyMatungaMumbai, MHIndia
| |
Collapse
|
21
|
An Invertebrate Host to Study Fungal Infections, Mycotoxins and Antifungal Drugs: Tenebrio molitor. J Fungi (Basel) 2018; 4:jof4040125. [PMID: 30424549 PMCID: PMC6308941 DOI: 10.3390/jof4040125] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/10/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
Faced with ethical conflict and social pressure, researchers have increasingly chosen to use alternative models over vertebrates in their research. Since the innate immune system is evolutionarily conserved in insects, the use of these animals in research is gaining ground. This review discusses Tenebrio molitor as a potential model host for the study of pathogenic fungi. Larvae of T. molitor are known as cereal pests and, in addition, are widely used as animal and human feed. A number of studies on mechanisms of the humoral system, especially in the synthesis of antimicrobial peptides, which have similar characteristics to vertebrates, have been performed. These studies demonstrate the potential of T. molitor larvae as a model host that can be used to study fungal virulence, mycotoxin effects, host immune responses to fungal infection, and the action of antifungal compounds.
Collapse
|
22
|
Yeh JJ, Lin CL, Hsu CY, Shae Z, Kao CH. Statin for Tuberculosis and Pneumonia in Patients with Asthma⁻Chronic Pulmonary Disease Overlap Syndrome: A Time-Dependent Population-Based Cohort Study. J Clin Med 2018; 7:E381. [PMID: 30355982 PMCID: PMC6262333 DOI: 10.3390/jcm7110381] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/04/2018] [Revised: 10/20/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022] Open
Abstract
We investigated the effects of statins on tuberculosis (TB) and pneumonia risks in asthma⁻chronic pulmonary disease overlap syndrome (ACOS) patients. We extracted data of patients diagnosed as having ACOS during 2000⁻2010 from the Taiwan National Health Insurance Research Database and divided them into statin users and nonusers. All study participants were followed up from the index date until death, withdrawal from insurance, or TB and pneumonia occurred (31 December 2011). The cumulative TB and pneumonia incidence was analyzed using Cox proportional regression analysis with time-dependent variables. After adjustments for multiple confounding factors including age, sex, comorbidities, and use of medications [statins, inhaled corticosteroids (ICSs), or oral steroids (OSs)], statin use was associated with significantly lower TB [adjusted hazard ratio (aHR) 0.49, 95% confidence interval (CI) 0.34⁻0.70] and pneumonia (aHR 0.52, 95% CI 0.41⁻0.65) risks. Moreover, aHRs (95% CIs) for statins combined with ICSs and OSs were respectively 0.60 (0.31⁻1.16) and 0.58 (0.40⁻0.85) for TB and 0.61 (0.39⁻0.95) and 0.57 (0.45⁻0.74) for pneumonia. Thus, statin users had lower TB and pneumonia risks than did nonusers, regardless of age, sex, comorbidities, and ICS or OS use. Pneumonia risk was lower among users of statins combined with ICSs or Oss and TB risk was lower among the users of statins combined with OSs.
Collapse
Affiliation(s)
- Jun-Jun Yeh
- Department of Family and Chest Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan.
- Department of Childhood Education and Nursery, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan.
- Department of Family Medicine, China Medical University, Taichung 40447, Taiwan.
- Department of Nursing, Mei-Ho University, Pingtung 91252, Taiwan.
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung 40447, Taiwan.
- College of Medicine, China Medical University, Taichung 40447, Taiwan.
| | - Chung-Y Hsu
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung 40447, Taiwan.
| | - Zonyin Shae
- Department of Computer Science and Information Engineering, Asia University, Taichung 40447, Taiwan.
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung 40447, Taiwan.
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung 40447, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 40447, Taiwan.
| |
Collapse
|
23
|
Kim H, Calatayud C, Guha S, Fernández-Carasa I, Berkowitz L, Carballo-Carbajal I, Ezquerra M, Fernández-Santiago R, Kapahi P, Raya Á, Miranda-Vizuete A, Lizcano JM, Vila M, Caldwell KA, Caldwell GA, Consiglio A, Dalfo E. The Small GTPase RAC1/CED-10 Is Essential in Maintaining Dopaminergic Neuron Function and Survival Against α-Synuclein-Induced Toxicity. Mol Neurobiol 2018; 55:7533-7552. [PMID: 29429047 PMCID: PMC6096980 DOI: 10.1007/s12035-018-0881-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/14/2017] [Accepted: 01/07/2018] [Indexed: 12/22/2022]
Abstract
Parkinson's disease is associated with intracellular α-synuclein accumulation and ventral midbrain dopaminergic neuronal death in the Substantia Nigra of brain patients. The Rho GTPase pathway, mainly linking surface receptors to the organization of the actin and microtubule cytoskeletons, has been suggested to participate to Parkinson's disease pathogenesis. Nevertheless, its exact contribution remains obscure. To unveil the participation of the Rho GTPase family to the molecular pathogenesis of Parkinson's disease, we first used C elegans to demonstrate the role of the small GTPase RAC1 (ced-10 in the worm) in maintaining dopaminergic function and survival in the presence of alpha-synuclein. In addition, ced-10 mutant worms determined an increase of alpha-synuclein inclusions in comparison to control worms as well as an increase in autophagic vesicles. We then used a human neuroblastoma cells (M17) stably over-expressing alpha-synuclein and found that RAC1 function decreased the amount of amyloidogenic alpha-synuclein. Further, by using dopaminergic neurons derived from patients of familial LRRK2-Parkinson's disease we report that human RAC1 activity is essential in the regulation of dopaminergic cell death, alpha-synuclein accumulation, participates in neurite arborization and modulates autophagy. Thus, we determined for the first time that RAC1/ced-10 participates in Parkinson's disease associated pathogenesis and established RAC1/ced-10 as a new candidate for further investigation of Parkinson's disease associated mechanisms, mainly focused on dopaminergic function and survival against α-synuclein-induced toxicity.
Collapse
Affiliation(s)
- Hanna Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Carles Calatayud
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain
- Center of Regenerative Medicine in Barcelona (CMRB), Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Hospital Duran i Reynals, 08908, L'Hospitalet de Llobregat, Spain
| | - Sanjib Guha
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Irene Fernández-Carasa
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain
| | - Laura Berkowitz
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Iria Carballo-Carbajal
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Mario Ezquerra
- Laboratory of Parkinson Disease and Other Neurodegenerative Movement Disorders, Department of Neurology: Clinical and Experimental Research, IDIBAPS - Hospital Clínic de Barcelona, 08036, Barcelona, Spain
| | - Rubén Fernández-Santiago
- Laboratory of Parkinson Disease and Other Neurodegenerative Movement Disorders, Department of Neurology: Clinical and Experimental Research, IDIBAPS - Hospital Clínic de Barcelona, 08036, Barcelona, Spain
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Ángel Raya
- Center of Regenerative Medicine in Barcelona (CMRB), Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Hospital Duran i Reynals, 08908, L'Hospitalet de Llobregat, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, 41013, Sevilla, Spain
| | - Jose Miguel Lizcano
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain.
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Spain.
| | - Esther Dalfo
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain.
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Can Baumann, 08500, Vic, Spain.
| |
Collapse
|
24
|
Arteaga Blanco LA, Crispim JS, Fernandes KM, de Oliveira LL, Pereira MF, Bazzolli DMS, Martins GF. Differential cellular immune response of Galleria mellonella to Actinobacillus pleuropneumoniae. Cell Tissue Res 2017; 370:153-168. [DOI: 10.1007/s00441-017-2653-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/17/2017] [Accepted: 05/30/2017] [Indexed: 11/25/2022]
|
25
|
Affiliation(s)
- Gholamreza Fazeli
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Ann Marie Wehman
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Vonk JJ, Yeshaw WM, Pinto F, Faber AIE, Lahaye LL, Kanon B, van der Zwaag M, Velayos-Baeza A, Freire R, van IJzendoorn SC, Grzeschik NA, Sibon OCM. Drosophila Vps13 Is Required for Protein Homeostasis in the Brain. PLoS One 2017; 12:e0170106. [PMID: 28107480 PMCID: PMC5249141 DOI: 10.1371/journal.pone.0170106] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/13/2016] [Accepted: 12/10/2016] [Indexed: 11/22/2022] Open
Abstract
Chorea-Acanthocytosis is a rare, neurodegenerative disorder characterized by progressive loss of locomotor and cognitive function. It is caused by loss of function mutations in the Vacuolar Protein Sorting 13A (VPS13A) gene, which is conserved from yeast to human. The consequences of VPS13A dysfunction in the nervous system are still largely unspecified. In order to study the consequences of VPS13A protein dysfunction in the ageing central nervous system we characterized a Drosophila melanogaster Vps13 mutant line. The Drosophila Vps13 gene encoded a protein of similar size as human VPS13A. Our data suggest that Vps13 is a peripheral membrane protein located to endosomal membranes and enriched in the fly head. Vps13 mutant flies showed a shortened life span and age associated neurodegeneration. Vps13 mutant flies were sensitive to proteotoxic stress and accumulated ubiquitylated proteins. Levels of Ref(2)P, the Drosophila orthologue of p62, were increased and protein aggregates accumulated in the central nervous system. Overexpression of the human Vps13A protein in the mutant flies partly rescued apparent phenotypes. This suggests a functional conservation of human VPS13A and Drosophila Vps13. Our results demonstrate that Vps13 is essential to maintain protein homeostasis in the larval and adult Drosophila brain. Drosophila Vps13 mutants are suitable to investigate the function of Vps13 in the brain, to identify genetic enhancers and suppressors and to screen for potential therapeutic targets for Chorea-Acanthocytosis.
Collapse
Affiliation(s)
- Jan J. Vonk
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wondwossen M. Yeshaw
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Francesco Pinto
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anita I. E. Faber
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Liza L. Lahaye
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart Kanon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marianne van der Zwaag
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Ofra s/n, La Laguna, Tenerife, Spain
| | - Sven C. van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nicola A. Grzeschik
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ody C. M. Sibon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
27
|
Zhou H, Yuan M, Yu Q, Zhou X, Min W, Gao D. Autophagy regulation and its role in gastric cancer and colorectal cancer. Cancer Biomark 2017; 17:1-10. [PMID: 27314289 DOI: 10.3233/cbm-160613] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Autophagy is associated with the occurrence, development, cellular adaptation, progression, treatment and prognosis of gastric cancer (GC) and colorectal cancer (CRC). The effect of autophagy in these two cancers has attracted our attention. OBJECTIVE The aim of this study was to describe the functional and regulatory mechanisms associated with autophagy in GC and CRC. METHODS We reviewed recent publications describing the role of autophagy in GC and CRC, including the functional characteristics, clinical significance and regulatory mechanisms. RESULTS Autophagy plays context-dependent dual roles in the development and progression of GC and CRC. It can either promote tumor growth and cell survival or can contribute to tumor suppression and promote cell death. Both of these effects employ complex regulatory networks, such as those mediated by p53, PI3K/Akt/mTOR, Ras and microRNA. Among the cellular process associated with these pathways, autophagy is a potential target for anti-tumor therapy. CONCLUSION Autophagy is associated with both tumorigenic and protective effects in cancer. However, the role of autophagy in GC and CRC remains unclear. Although the translation of the basic science of autophagy into clinical practice is a long process, the modulation of autophagy as a potential therapeutic approach in GC and CRC merits further investigation.
Collapse
Affiliation(s)
- Huangyan Zhou
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Academy of Medical Sciences, Nanchang, Jiangxi, China.,Institute of Immunotherapy, Nanchang University, Nanchang, Jiangxi, China
| | - Min Yuan
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyan Zhou
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Weiping Min
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Academy of Medical Sciences, Nanchang, Jiangxi, China.,Institute of Immunotherapy, Nanchang University, Nanchang, Jiangxi, China
| | - Dian Gao
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Academy of Medical Sciences, Nanchang, Jiangxi, China.,Institute of Immunotherapy, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
28
|
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been implicated in a wide range of cellular processes, including the catabolic pathways collectively described as autophagy. In this chapter, the evidence linking LRRK2 to autophagy will be examined, along with how regulation of autophagy and lysosomal pathways may provide a nexus between the physiological function of this protein and the different diseases with which it has been associated. Data from cellular and animal models for LRRK2 function and dysfunction support a role in the regulation and control of autophagic pathways in the cell, although the extant results do not provide a clear indication as to whether LRRK2 is a positive or negative regulator of these pathways, and there are conflicting data as to the impact of mutations in LRRK2 causative for Parkinson's disease. Given that LRRK2 is a priority drug target for Parkinson's, the evidence suggesting that knockout or inhibition of LRRK2 can result in deregulation of autophagy may have important implications and is discussed in the context of our wider understanding of LRRK2.
Collapse
Affiliation(s)
- Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK.
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|
29
|
Gallorini M, di Giacomo V, Di Valerio V, Rapino M, Bosco D, Travan A, Di Giulio M, Di Pietro R, Paoletti S, Cataldi A, Sancilio S. Cell-protection mechanism through autophagy in HGFs/S. mitis co-culture treated with Chitlac-nAg. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:186. [PMID: 27787811 DOI: 10.1007/s10856-016-5803-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/21/2016] [Accepted: 10/14/2016] [Indexed: 02/08/2023]
Abstract
Silver-based products have been proven to be effective in retarding and preventing bacterial growth since ancient times. In the field of restorative dentistry, the use of silver ions/nanoparticles has been explored to counteract bacterial infections, as silver can destroy bacterial cell walls by reacting with membrane proteins. However, it is also cytotoxic towards eukaryotic cells, which are capable of internalizing nanoparticles. In this work, we investigated the biological effects of Chitlac-nAg, a colloidal system based on a modified chitosan (Chitlac), administered for 24-48 h to a co-culture of primary human gingival fibroblasts and Streptococcus mitis in the presence of saliva, developed to mimic the microenvironment of the oral cavity. We sought to determine its efficiency to combat oral hygiene-related diseases without affecting eukaryotic cells. Cytotoxicity, reactive oxygen species production, apoptosis induction, nanoparticles uptake, and lysosome and autophagosome metabolism were evaluated. In vitro results show that Chitlac-nAg does not exert cytotoxic effects on human gingival fibroblasts, which seem to survive through a homoeostasis mechanism involving autophagy. That suggests that the novel biomaterial Chitlac-nAg could be a promising tool in the field of dentistry.
Collapse
Affiliation(s)
- Marialucia Gallorini
- Department of Pharmacy, "G. d'Annunzio" University, Chieti-Pescara, Italy
- Department of Operative Dentistry and Periodontology, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University, Chieti-Pescara, Italy.
| | - Valentina Di Valerio
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Monica Rapino
- Genetic Molecular Institute of CNR, Unit of Chieti, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Domenico Bosco
- Genetic Molecular Institute of CNR, Unit of Chieti, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Andrea Travan
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Mara Di Giulio
- Department of Pharmacy, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Sergio Paoletti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Amelia Cataldi
- Department of Pharmacy, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Silvia Sancilio
- Department of Pharmacy, "G. d'Annunzio" University, Chieti-Pescara, Italy
| |
Collapse
|
30
|
Sex-specific pharmacological modulation of autophagic process in human umbilical artery smooth muscle cells. Pharmacol Res 2016; 113:166-174. [DOI: 10.1016/j.phrs.2016.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/03/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 01/29/2023]
|
31
|
Immune tolerance to an intestine-adapted bacteria, Chryseobacterium sp., injected into the hemocoel of Protaetia brevitarsis seulensis. Sci Rep 2016; 6:31722. [PMID: 27530146 PMCID: PMC4987663 DOI: 10.1038/srep31722] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/09/2016] [Accepted: 07/22/2016] [Indexed: 11/11/2022] Open
Abstract
To explore the interaction of gut microbes and the host immune system, bacteria were isolated from the gut of Protaetia brevitarsis seulensis larvae. Chryseobacterium sp., Bacillus subtilis, Arthrobacter arilaitensis, Bacillus amyloliquefaciens, Bacillus megaterium, and Lysinibacillus xylanilyticus were cultured in vitro, identified, and injected in the hemocoel of P. brevitarsis seulensis larvae, respectively. There were no significant changes in phagocytosis-associated lysosomal formation or pathogen-related autophagosome in immune cells (granulocytes) from Chryseobacterium sp.-challenged larvae. Next, we examined changes in the transcription of innate immune genes such as peptidoglycan recognition proteins and antimicrobial peptides following infection with Chryseobacterium sp. PGRP-1 and -2 transcripts, which may be associated with melanization generated by prophenoloxidase (PPO), were either highly or moderately expressed at 24 h post-infection with Chryseobacterium sp. However, PGRP-SC2 transcripts, which code for bactericidal amidases, were expressed at low levels. With respect to antimicrobial peptides, only coleoptericin was moderately expressed in Chryseobacterium sp.-challenged larvae, suggesting maintenance of an optimum number of Chryseobacterium sp. All examined genes were expressed at significantly higher levels in larvae challenged with a pathogenic bacterium. Our data demonstrated that gut-inhabiting bacteria, the Chryseobacterium sp., induced a weaker immune response than other pathogenic bacteria, E. coli K12.
Collapse
|
32
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
33
|
Abstract
Many common renal insults such as ischemia and toxic injury primarily target the tubular epithelial cells, especially the highly metabolically active proximal tubular segment. Tubular epithelial cells are particularly dependent on autophagy to maintain homeostasis and respond to stressors. The pattern of autophagy in the kidney has a unique spatial and chronologic signature. Recent evidence has shown that there is complex cross-talk between autophagy and various cell death pathways. This review specifically discusses the interplay between autophagy and cell death in the renal tubular epithelia. It is imperative to review this topic because recent discoveries have improved our mechanistic understanding of the autophagic process and have highlighted its broad clinical applications, making autophagy a major target for drug development.
Collapse
Affiliation(s)
- Andrea Havasi
- Department of Nephrology, Boston University Medical Center, Boston, MA.
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia and Charlie Norwood VA Medical Center, Augusta, GA
| |
Collapse
|
34
|
Brooks CR, Yeung MY, Brooks YS, Chen H, Ichimura T, Henderson JM, Bonventre JV. KIM-1-/TIM-1-mediated phagocytosis links ATG5-/ULK1-dependent clearance of apoptotic cells to antigen presentation. EMBO J 2015; 34:2441-64. [PMID: 26282792 DOI: 10.15252/embj.201489838] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2014] [Accepted: 07/01/2015] [Indexed: 12/14/2022] Open
Abstract
Phagocytosis of apoptotic cells by both professional and semi-professional phagocytes is required for resolution of organ damage and maintenance of immune tolerance. KIM-1/TIM-1 is a phosphatidylserine receptor that is expressed on epithelial cells and can transform the cells into phagocytes. Here, we demonstrate that KIM-1 phosphorylation and association with p85 results in encapsulation of phagosomes by lipidated LC3 in multi-membrane organelles. KIM-1-mediated phagocytosis is not associated with increased ROS production, and NOX inhibition does not block LC3 lipidation. Autophagy gene expression is required for efficient clearance of apoptotic cells and phagosome maturation. KIM-1-mediated phagocytosis leads to pro-tolerogenic antigen presentation, which suppresses CD4 T-cell proliferation and increases the percentage of regulatory T cells in an autophagy gene-dependent manner. Taken together, these data reveal a novel mechanism of epithelial biology linking phagocytosis, autophagy and antigen presentation to regulation of the inflammatory response.
Collapse
Affiliation(s)
- Craig R Brooks
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Transplantation Research Center, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Yang S Brooks
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA, USA Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Takaharu Ichimura
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Joseph V Bonventre
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
35
|
Santos Franco S, Raveh-Amit H, Kobolák J, Alqahtani MH, Mobasheri A, Dinnyes A. The crossroads between cancer stem cells and aging. BMC Cancer 2015; 15 Suppl 1:S1. [PMID: 25708542 PMCID: PMC4331724 DOI: 10.1186/1471-2407-15-s1-s1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis suggests that only a subpopulation of cells within a tumour is responsible for the initiation and progression of neoplasia. The original and best evidence for the existence of CSCs came from advances in the field of haematological malignancies. Thus far, putative CSCs have been isolated from various solid and non-solid tumours and shown to possess self-renewal, differentiation, and cancer regeneration properties. Although research in the field is progressing extremely fast, proof of concept for the CSC hypothesis is still lacking and key questions remain unanswered, e.g. the cell of origin for these cells. Nevertheless, it is undisputed that neoplastic transformation is associated with genetic and epigenetic alterations of normal cells, and a better understanding of these complex processes is of utmost importance for developing new anti-cancer therapies. In the present review, we discuss the CSC hypothesis with special emphasis on age-associated alterations that govern carcinogenesis, at least in some types of tumours. We present evidence from the scientific literature for age-related genetic and epigenetic alterations leading to cancer and discuss the main challenges in the field.
Collapse
|
36
|
Frost LS, Lopes VS, Bragin A, Reyes-Reveles J, Brancato J, Cohen A, Mitchell CH, Williams DS, Boesze-Battaglia K. The Contribution of Melanoregulin to Microtubule-Associated Protein 1 Light Chain 3 (LC3) Associated Phagocytosis in Retinal Pigment Epithelium. Mol Neurobiol 2014; 52:1135-1151. [PMID: 25301234 DOI: 10.1007/s12035-014-8920-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2014] [Accepted: 09/29/2014] [Indexed: 02/07/2023]
Abstract
A main requisite in the phagocytosis of ingested material is a coordinated series of maturation steps which lead to the degradation of ingested cargo. Photoreceptor outer segment (POS) renewal involves phagocytosis of the distal disk membranes by the retinal pigment epithelium (RPE). Previously, we identified melanoregulin (MREG) as an intracellular cargo-sorting protein required for the degradation of POS disks. Here, we provide evidence that MREG-dependent processing links both autophagic and phagocytic processes in LC3-associated phagocytosis (LAP). Ingested POS phagosomes are associated with endogenous LC3 and MREG. The LC3 association with POSs exhibited properties of LAP; it was independent of rapamycin pretreatment, but dependent on Atg5. Loss of MREG resulted in a decrease in the extent of LC3-POS association. Studies using DQ-BSA suggest that loss of MREG does not compromise the association and fusion of LC3-positive phagosomes with lysosomes. Furthermore, the mechanism of MREG action is likely through a protein complex that includes LC3, as determined by colocalization and immunoprecipitation in both RPE cells and macrophages. We posit that MREG participates in coordinating the association of phagosomes with LC3 for content degradation with the loss of MREG leading to phagosome accumulation.
Collapse
Affiliation(s)
- Laura S Frost
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vanda S Lopes
- UCLA School of Medicine, Jules Stein Eye Institute, Los Angeles, CA, 90095, USA
| | - Alvina Bragin
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Juan Reyes-Reveles
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jennifer Brancato
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Art Cohen
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David S Williams
- UCLA School of Medicine, Jules Stein Eye Institute, Los Angeles, CA, 90095, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Kwon H, Bang K, Cho S. Characterization of the hemocytes in Larvae of Protaetia brevitarsis seulensis: involvement of granulocyte-mediated phagocytosis. PLoS One 2014; 9:e103620. [PMID: 25083702 PMCID: PMC4118905 DOI: 10.1371/journal.pone.0103620] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2014] [Accepted: 06/29/2014] [Indexed: 12/18/2022] Open
Abstract
Hemocytes are key players in the immune response against pathogens in insects. However, the hemocyte types and their functions in the white-spotted flower chafers, Protaetia brevitarsis seulensis (Kolbe), are not known. In this study, we used various microscopes, molecular probes, and flow cytometric analyses to characterize the hemocytes in P. brevitarsis seulensis. The circulating hemocytes were classified based on their size, morphology, and dye-staining properties into six types, including granulocytes, plasmatocytes, oenocytoids, spherulocytes, prohemocytes, and adipohemocytes. The percentages of circulating hemocyte types were as follows: 13% granulocytes, 20% plasmatocytes, 1% oenocytoids, 5% spherulocytes, 17% prohemocytes, and 44% adipohemocytes. Next, we identified the professional phagocytes, granulocytes, which mediate encapsulation and phagocytosis of pathogens. The granulocytes were immunologically or morphologically activated and phagocytosed potentially hazardous substances in vivo. In addition, we showed that the phagocytosis by granulocytes is associated with autophagy, and that the activation of autophagy could be an efficient way to eliminate pathogens in this system. We also observed a high accumulation of autophagic vacuoles in activated granulocytes, which altered their shape and led to autophagic cell death. Finally, the granulocytes underwent mitotic division thus maintaining their number in vivo.
Collapse
Affiliation(s)
- Hyojung Kwon
- Department of Applied Biology, College of Agriculture and Life Science, Environment Friendly Agriculture Center, Kangwon National University, Chuncheon, Republic of Korea
| | - Kyeongrin Bang
- Department of Applied Biology, College of Agriculture and Life Science, Environment Friendly Agriculture Center, Kangwon National University, Chuncheon, Republic of Korea
| | - Saeyoull Cho
- Department of Applied Biology, College of Agriculture and Life Science, Environment Friendly Agriculture Center, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
38
|
Liang Z, Zhang Q, Thomas CM, Chana KK, Gibeon D, Barnes PJ, Chung KF, Bhavsar PK, Donnelly LE. Impaired macrophage phagocytosis of bacteria in severe asthma. Respir Res 2014; 15:72. [PMID: 24972601 PMCID: PMC4086996 DOI: 10.1186/1465-9921-15-72] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2014] [Accepted: 06/20/2014] [Indexed: 01/18/2023] Open
Abstract
Background Bacteria are frequently cultured from sputum samples of severe asthma patients suggesting a defect in bacterial clearance from the airway. We measured the capacity of macrophages from patients with asthma to phagocytose bacteria. Methods Phagocytosis of fluorescently-labelled polystyrene beads, Haemophilus influenzae or Staphylococcus aureus by broncholaveolar lavage alveolar macrophages (AM) and by monocyte-derived macrophages (MDM) from non-asthmatics, mild-moderate and severe asthmatic patients was assessed using fluorimetry. Results There were no differences in phagocytosis of polystyrene beads by AMs or MDMs from any of the subject groups. There was reduced phagocytosis of Haemophilus influenzae and Staphylococcus aureus in MDMs from patients with severe asthma compared to non-severe asthma (p < 0.05 and p < 0.01, respectively) and healthy subjects (p < 0.01and p < 0.001, respectively). Phagocytosis of Haemophilus influenzae and Staphylococcus aureus by AM was also reduced in severe asthma compared to normal subjects (p < 0.05). Dexamethasone and formoterol did not suppress phagocytosis of bacteria by MDMs from any of the groups. Conclusions Persistence of bacteria in the lower airways may result partly from a reduced phagocytic capacity of macrophages for bacteria. This may contribute to increased exacerbations, airway colonization and persistence of inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pankaj K Bhavsar
- Airway Disease, National Heart and Lung Institute, Imperial College London, & Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London SW3, UK.
| | | |
Collapse
|
39
|
Frost LS, Mitchell CH, Boesze-Battaglia K. Autophagy in the eye: implications for ocular cell health. Exp Eye Res 2014; 124:56-66. [PMID: 24810222 DOI: 10.1016/j.exer.2014.04.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/21/2014] [Revised: 04/10/2014] [Accepted: 04/12/2014] [Indexed: 12/21/2022]
Abstract
Autophagy, a catabolic process by which a cell "eats" itself, turning over its own cellular constituents, plays a key role in cellular homeostasis. In an effort to maintain normal cellular function, autophagy is often up-regulated in response to environmental stresses and excessive organelle damage to facilitate aggregated protein removal. In the eye, virtually all cell types from those comprising the cornea in the front of the eye to the retinal pigment epithelium (RPE) providing a protective barrier for the retina at the back of the eye, rely on one or more aspects of autophagy to maintain structure and/or normal physiological function. In the lens autophagy plays a critical role in lens fiber cell maturation and the formation of the organelle free zone. Numerous studies delineating the role of Atg5, Vsp34 as well as FYCO1 in maintenance of lens transparency are discussed. Corneal endothelial dystrophies are also characterized as having elevated levels of autophagic proteins. Therefore, novel modulators of autophagy such as lithium and melatonin are proposed as new therapeutic strategies for this group of dystrophies. In addition, we summarize how corneal Herpes Simplex Virus (HSV-1) infection subverts the cornea's response to infection by inhibiting the normal autophagic response. Using glaucoma models we analyze the relative contribution of autophagy to cell death and cell survival. The cytoprotective role of autophagy is further discussed in an analysis of photoreceptor cell heath and function. We focus our analysis on the current understanding of autophagy in photoreceptor and RPE health, specifically on the diverse role of autophagy in rods and cones as well as its protective role in light induced degeneration. Lastly, in the RPE we highlight hybrid phagocytosis-autophagy pathways. This comprehensive review allows us to speculate on how alterations in various stages of autophagy contribute to glaucoma and retinal degenerations.
Collapse
Affiliation(s)
- Laura S Frost
- Department of Biochemistry, University of Pennsylvania, SDM, Philadelphia, PA 19104, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, SDM, Philadelphia, PA 19104, USA
| | | |
Collapse
|
40
|
Protective effect of astaxanthin on liver fibrosis through modulation of TGF-β1 expression and autophagy. Mediators Inflamm 2014; 2014:954502. [PMID: 24860243 PMCID: PMC4016904 DOI: 10.1155/2014/954502] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/04/2013] [Revised: 02/16/2014] [Accepted: 02/25/2014] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a common pathway leading to cirrhosis and a worldwide clinical issue. Astaxanthin is a red carotenoid pigment with antioxidant, anticancer, and anti-inflammatory properties. The aim of this study was to investigate the effect of astaxanthin on liver fibrosis and its potential protective mechanisms. Liver fibrosis was induced in a mouse model using CCL4 (intraperitoneal injection, three times a week for 8 weeks), and astaxanthin was administered everyday at three doses (20, 40, and 80 mg/kg). Pathological results indicated that astaxanthin significantly improved the pathological lesions of liver fibrosis. The levels of alanine aminotransferase aspartate aminotransferase and hydroxyproline were also significantly decreased by astaxanthin. The same results were confirmed in bile duct liagtion, (BDL) model. In addition, astaxanthin inhibited hepatic stellate cells (HSCs) activation and formation of extracellular matrix (ECM) by decreasing the expression of NF-κB and TGF-β1 and maintaining the balance between MMP2 and TIMP1. In addition, astaxanthin reduced energy production in HSCs by downregulating the level of autophagy. These results were simultaneously confirmed in vivo and in vitro. In conclusion, our study showed that 80 mg/kg astaxanthin had a significant protective effect on liver fibrosis by suppressing multiple profibrogenic factors.
Collapse
|
41
|
Valapala M, Wilson C, Hose S, Bhutto IA, Grebe R, Dong A, Greenbaum S, Gu L, Sengupta S, Cano M, Hackett S, Xu G, Lutty GA, Dong L, Sergeev Y, Handa JT, Campochiaro P, Wawrousek E, Zigler JS, Sinha D. Lysosomal-mediated waste clearance in retinal pigment epithelial cells is regulated by CRYBA1/βA3/A1-crystallin via V-ATPase-MTORC1 signaling. Autophagy 2014; 10:480-96. [PMID: 24468901 PMCID: PMC4077886 DOI: 10.4161/auto.27292] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022] Open
Abstract
In phagocytic cells, including the retinal pigment epithelium (RPE), acidic compartments of the endolysosomal system are regulators of both phagocytosis and autophagy, thereby helping to maintain cellular homeostasis. The acidification of the endolysosomal system is modulated by a proton pump, the V-ATPase, but the mechanisms that direct the activity of the V-ATPase remain elusive. We found that in RPE cells, CRYBA1/βA3/A1-crystallin, a lens protein also expressed in RPE, is localized to lysosomes, where it regulates endolysosomal acidification by modulating the V-ATPase, thereby controlling both phagocytosis and autophagy. We demonstrated that CRYBA1 coimmunoprecipitates with the ATP6V0A1/V0-ATPase a1 subunit. Interestingly, in mice when Cryba1 (the gene encoding both the βA3- and βA1-crystallin forms) is knocked out specifically in RPE, V-ATPase activity is decreased and lysosomal pH is elevated, while cathepsin D (CTSD) activity is decreased. Fundus photographs of these Cryba1 conditional knockout (cKO) mice showed scattered lesions by 4 months of age that increased in older mice, with accumulation of lipid-droplets as determined by immunohistochemistry. Transmission electron microscopy (TEM) of cryba1 cKO mice revealed vacuole-like structures with partially degraded cellular organelles, undigested photoreceptor outer segments and accumulation of autophagosomes. Further, following autophagy induction both in vivo and in vitro, phospho-AKT and phospho-RPTOR/Raptor decrease, while pMTOR increases in RPE cells, inhibiting autophagy and AKT-MTORC1 signaling. Impaired lysosomal clearance in the RPE of the cryba1 cKO mice also resulted in abnormalities in retinal function that increased with age, as demonstrated by electroretinography. Our findings suggest that loss of CRYBA1 causes lysosomal dysregulation leading to the impairment of both autophagy and phagocytosis.
Collapse
Affiliation(s)
- Mallika Valapala
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Christine Wilson
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Stacey Hose
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Imran A Bhutto
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Rhonda Grebe
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Aling Dong
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Seth Greenbaum
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Limin Gu
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA; Department of Ophthalmology of Shanghai Tenth People's Hospital and Tongji Eye Institute; Tongji University School of Medicine; Shanghai, China
| | - Samhita Sengupta
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Marisol Cano
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Sean Hackett
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Guotong Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital and Tongji Eye Institute; Tongji University School of Medicine; Shanghai, China
| | - Gerard A Lutty
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Lijin Dong
- National Eye Institute; National Institutes of Health; Bethesda, MD USA
| | - Yuri Sergeev
- National Eye Institute; National Institutes of Health; Bethesda, MD USA
| | - James T Handa
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Peter Campochiaro
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Eric Wawrousek
- National Eye Institute; National Institutes of Health; Bethesda, MD USA
| | - J Samuel Zigler
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Debasish Sinha
- Wilmer Eye Institute; Johns Hopkins University School of Medicine; Baltimore, MD USA
| |
Collapse
|
42
|
Mehla K, Singh PK. MUC1: a novel metabolic master regulator. Biochim Biophys Acta Rev Cancer 2014; 1845:126-35. [PMID: 24418575 DOI: 10.1016/j.bbcan.2014.01.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/25/2013] [Revised: 12/28/2013] [Accepted: 01/03/2014] [Indexed: 12/14/2022]
Abstract
MUC1, a type I transmembrane protein, is significantly overexpressed and aberrantly glycosylated in tumors of epithelial origin. By virtue of its aberrant signaling due to loss of apical-basal polarity in cancer, MUC1 regulates the metabolite flux at multiple levels. Serving as a transcriptional co-activator, MUC1 directly regulates expression of metabolic genes. By regulating receptor tyrosine kinase signaling, MUC1 facilitates production of biosynthetic intermediates required for cell growth. Also, via direct interactions, MUC1 modulates the activity/stability of enzymes and transcription factors that directly regulate metabolic functions. Additionally, by modulation of autophagy, levels of reactive oxygen species, and metabolite flux, MUC1 facilitates cancer cell survival under hypoxic and nutrient-deprived conditions. This article provides a comprehensive review of recent literature on novel metabolic functions of MUC1.
Collapse
Affiliation(s)
- Kamiya Mehla
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pankaj K Singh
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Genetic Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
43
|
Vural A, Kehrl JH. Autophagy in macrophages: impacting inflammation and bacterial infection. SCIENTIFICA 2014; 2014:825463. [PMID: 24818040 PMCID: PMC4000662 DOI: 10.1155/2014/825463] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 01/15/2014] [Accepted: 02/28/2014] [Indexed: 05/09/2023]
Abstract
Macrophages are on the front line of host defense. They possess an array of germline-encoded pattern recognition receptors/sensors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) and which activate downstream effectors/pathways to help mediate innate immune responses and host defense. Innate immune responses include the rapid induction of transcriptional networks that trigger the production of cytokines, chemokines, and cytotoxic molecules; the mobilization of cells including neutrophils and other leukocytes; the engulfment of pathogens by phagocytosis and their delivery to lysosome for degradation; and the induction of autophagy. Autophagy is a catabolic process that normally maintains cellular homeostasis in a lysosome-dependent manner, but it also functions as a cytoprotective response that intersects with a variety of general stress-response pathways. This review focuses on the intimately linked molecular mechanisms that help govern the autophagic pathway and macrophage innate immune responses.
Collapse
Affiliation(s)
- Ali Vural
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 11N214, Center Drive, MSC 1876, Bethesda, MD 20892, USA
| | - John H. Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 11N214, Center Drive, MSC 1876, Bethesda, MD 20892, USA
- *John H. Kehrl:
| |
Collapse
|
44
|
Verma M, Steer EK, Chu CT. ERKed by LRRK2: a cell biological perspective on hereditary and sporadic Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2013; 1842:1273-81. [PMID: 24225420 DOI: 10.1016/j.bbadis.2013.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/31/2013] [Revised: 10/14/2013] [Accepted: 11/03/2013] [Indexed: 02/08/2023]
Abstract
The leucine rich repeat kinase 2 (LRRK2/dardarin) is implicated in autosomal dominant familial and sporadic Parkinson's disease (PD); mutations in LRRK2 account for up to 40% of PD cases in some populations. LRRK2 is a large protein with a kinase domain, a GTPase domain, and multiple potential protein interaction domains. As such, delineating the functional pathways for LRRK2 and mechanisms by which PD-linked variants contribute to age-related neurodegeneration could result in pharmaceutically tractable therapies. A growing number of recent studies implicate dysregulation of mitogen activated protein kinases 3 and 1 (also known as ERK1/2) as possible downstream mediators of mutant LRRK2 effects. As these master regulators of growth, differentiation, neuronal plasticity and cell survival have also been implicated in other PD models, a set of common cell biological pathways may contribute to neuronal susceptibility in PD. Here, we review the literature on several major cellular pathways impacted by LRRK2 mutations--autophagy, microtubule/cytoskeletal dynamics, and protein synthesis--in context of potential signaling crosstalk involving the ERK1/2 and Wnt signaling pathways. Emerging implications for calcium homeostasis, mitochondrial biology and synaptic dysregulation are discussed in relation to LRRK2 interactions with other PD gene products. It has been shown that substantia nigra neurons in human PD and Lewy body dementia patients exhibit cytoplasmic accumulations of ERK1/2 in mitochondria, autophagosomes and bundles of intracellular fibrils. Both experimental and human tissue data implicate pathogenic changes in ERK1/2 signaling in sporadic, toxin-based and mutant LRRK2 settings, suggesting engagement of common cell biological pathways by divergent PD etiologies.
Collapse
Affiliation(s)
- Manish Verma
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Erin K Steer
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Charleen T Chu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
45
|
Survival or death: disequilibrating the oncogenic and tumor suppressive autophagy in cancer. Cell Death Dis 2013; 4:e892. [PMID: 24176850 PMCID: PMC3920945 DOI: 10.1038/cddis.2013.422] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2013] [Revised: 09/20/2013] [Accepted: 09/23/2013] [Indexed: 02/05/2023]
Abstract
Autophagy (macroautophagy) is an evolutionarily conserved lysosomal degradation process, in which a cell degrades long-lived proteins and damaged organelles. Recently, accumulating evidence has revealed the core molecular machinery of autophagy in carcinogenesis; however, the intricate relationship between autophagy and cancer continue to remain an enigma. Why does autophagy have either pro-survival (oncogenic) or pro-death (tumor suppressive) role at different cancer stages, including cancer stem cell, initiation and progression, invasion and metastasis, as well as dormancy? How does autophagy modulate a series of oncogenic and/or tumor suppressive pathways, implicated in microRNA (miRNA) involvement? Whether would targeting the oncogenic and tumor suppressive autophagic network be a novel strategy for drug discovery? To address these problems, we focus on summarizing the dynamic oncogenic and tumor suppressive roles of autophagy and their relevant small-molecule drugs, which would provide a new clue to elucidate the oncosuppressive (survival or death) autophagic network as a potential therapeutic target.
Collapse
|
46
|
Nelson MP, Shacka JJ. Autophagy Modulation in Disease Therapy: Where Do We Stand? CURRENT PATHOBIOLOGY REPORTS 2013; 1:239-245. [PMID: 24470989 DOI: 10.1007/s40139-013-0032-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022]
Abstract
Since it was first described more than 50 years ago autophagy has been examined in many contexts, from cell survival to pathogen sequestration and removal. In more recent years our understanding of autophagy has developed sufficiently to allow effective targeted therapeutics to be developed against various diseases. The field of autophagy research is expanding rapidly, demonstrated by increases in both numbers of investigators in the field and the breadth of topics being addressed. Some diseases, such as the many cancers, have come to the fore in autophagy therapeutics research as a better understanding of their underlying mechanisms has surfaced. Numerous treatments are being developed and explored, from creative applications of the classic autophagy modulators chloroquine and rapamycin, to repurposing drugs approved for other treatments, such as astemizole, which is currently in use as an antimalarial and chronic rhinitis treatment. The landscape of autophagy modulation in disease therapy is rapidly changing and this review hopes to provide a cross-section of the current state of the field.
Collapse
Affiliation(s)
- Michael P Nelson
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| | - John J Shacka
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham VA Medical Center, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| |
Collapse
|
47
|
Forestieri R, Donohue E, Balgi A, Roberge M, Andersen RJ. Synthesis of clionamine B, an autophagy stimulating aminosteroid isolated from the sponge Cliona celata. Org Lett 2013; 15:3918-21. [PMID: 23869546 DOI: 10.1021/ol4016783] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/01/2023]
Abstract
Clionamine B (2), an aminosteroid isolated from the marine sponge Cliona celata, has been synthesized starting from the plant sapogenin tigogenin (5). A key step in the synsthesis is the stereoselective introduction of the C-20 α-hydroxyl substituent via oxidation of a γ-lactone enolate with molecular oxygen. Synthetic clionamine B (2) strongly stimulated autophagy in human breast cancer MCF-7 cells.
Collapse
Affiliation(s)
- Roberto Forestieri
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B. C., Canada, V6T 1Z1
| | | | | | | | | |
Collapse
|