1
|
Moreno-Blas D, Adell T, González-Estévez C. Autophagy in Tissue Repair and Regeneration. Cells 2025; 14:282. [PMID: 39996754 PMCID: PMC11853389 DOI: 10.3390/cells14040282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/01/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy is a cellular recycling system that, through the sequestration and degradation of intracellular components regulates multiple cellular functions to maintain cellular homeostasis and survival. Dysregulation of autophagy is closely associated with the development of physiological alterations and human diseases, including the loss of regenerative capacity. Tissue regeneration is a highly complex process that relies on the coordinated interplay of several cellular processes, such as injury sensing, defense responses, cell proliferation, differentiation, migration, and cellular senescence. These processes act synergistically to repair or replace damaged tissues and restore their morphology and function. In this review, we examine the evidence supporting the involvement of the autophagy pathway in the different cellular mechanisms comprising the processes of regeneration and repair across different regenerative contexts. Additionally, we explore how modulating autophagy can enhance or accelerate regeneration and repair, highlighting autophagy as a promising therapeutic target in regenerative medicine for the development of autophagy-based treatments for human diseases.
Collapse
Affiliation(s)
| | | | - Cristina González-Estévez
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal, 643, 08028 Barcelona, Spain; (D.M.-B.); (T.A.)
| |
Collapse
|
2
|
Zhu Y, Tan J, Wang Y, Gong Y, Zhang X, Yuan Z, Lu X, Tang H, Zhang Z, Jiang X, Zhu W, Gong L. Atg5 deficiency in macrophages protects against kidney fibrosis via the CCR6-CCL20 axis. Cell Commun Signal 2024; 22:223. [PMID: 38594728 PMCID: PMC11003172 DOI: 10.1186/s12964-024-01600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Autophagy is a lysosome-dependent degradation pathway that regulates macrophage activation, differentiation, and polarization. Autophagy related 5 (Atg5) is a key protein involved in phagocytic membrane elongation in autophagic vesicles that forms a complex with Atg12 and Atg16L1. Alterations in Atg5 are related to both acute and chronic kidney diseases in experimental models. However, the role of macrophage-expressed Atg5 in acute kidney injury remains unclear. METHODS Using a myeloid cell-specific Atg5 knockout (MΦ atg5-/-) mouse, we established renal ischemia/reperfusion and unilateral ureteral obstruction models to evaluate the role of macrophage Atg5 in renal macrophage migration and fibrosis. RESULTS Based on changes in the serum urea nitrogen and creatinine levels, Atg5 deletion had a minimal effect on renal function in the early stages after mild injury; however, MΦ atg5-/- mice had reduced renal fibrosis and reduced macrophage recruitment after 4 weeks of ischemia/reperfusion injury and 2 weeks of unilateral ureteral obstruction injury. Atg5 deficiency impaired the CCL20-CCR6 axis after severe ischemic kidneys. Chemotactic responses of bone marrow-derived monocytes (BMDMs) from MΦ atg5-/- mice to CCL20 were significantly attenuated compared with those of wild-type BMDMs, and this might be caused by the inhibition of PI3K, AKT, and ERK1/2 activation. CONCLUSIONS Our data indicate that Atg5 deficiency decreased macrophage migration by impairing the CCL20-CCR6 axis and inhibited M2 polarization, thereby improving kidney fibrosis.
Collapse
Affiliation(s)
- Yufeng Zhu
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, China
| | - Jiexing Tan
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, China
| | - Yuanzhan Wang
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, China
| | - Yuhong Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziguo Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinyu Lu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huifang Tang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Zhiming Zhang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Proteomics, Guangzhou, China
| | - Wei Zhu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Jobst M, Hossain M, Kiss E, Bergen J, Marko D, Del Favero G. Autophagy modulation changes mechano-chemical sensitivity of T24 bladder cancer cells. Biomed Pharmacother 2024; 170:115942. [PMID: 38042111 DOI: 10.1016/j.biopha.2023.115942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/27/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
Bladder cancer cells possess unique adaptive capabilities: shaped by their environment, cells face a complex chemical mixture of metabolites and xenobiotics accompanied by physiological mechanical cues. These responses might translate into resistance to chemotherapeutical regimens and can largely rely on autophagy. Considering molecules capable of rewiring tumor plasticity, compounds of natural origin promise to offer valuable options. Fungal derived metabolites, such as bafilomycin and wortmannin are widely acknowledged as autophagy inhibitors. Here, their potential to tune bladder cancer cells´ adaptability to chemical and physical stimuli was assessed. Additionally, dietary occurring mycotoxins were also investigated, namely deoxynivalenol (DON, 0.1-10 µM) and fusaric acid (FA, 0.1-1 mM). Endowing a Janus' face behavior, DON and FA are on the one side described as toxins with detrimental health effects. Concomitantly, they are also explored experimentally for selective pharmacological applications including anticancer activities. In non-cytotoxic concentrations, bafilomycin (BAFI, 1-10 nM) and wortmannin (WORT, 1 µM) modified cell morphology and reduced cancer cell migration. Application of shear stress and inhibition of mechano-gated PIEZO channels reduced cellular sensitivity to BAFI treatment (1 nM). Similarly, for FA (0.5 mM) PIEZO1 expression and inhibition largely aligned with the modulatory potential on cancer cells motility. Additionally, this study highlighted that the activity profile of compounds with similar cytotoxic potential (e.g. co-incubation DON with BAFI or FA with WORT) can diverge substantially in the regulation of cell mechanotransduction. Considering the interdependence between tumor progression and response to mechanical cues, these data promise to provide a novel viewpoint for the study of chemoresistance and associated pathways.
Collapse
Affiliation(s)
- Maximilian Jobst
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Maliha Hossain
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Janice Bergen
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria.
| |
Collapse
|
4
|
Yang Y, Jiang S, Yang J, Feng X, Wang C, Wang K, Gao W, Du X, Lei L, Wang Z, Liu G, Song Y, Li X. β-hydroxybutyrate impairs the directionality of migrating neutrophils through inhibiting the autophagy-dependent degradation of Cdc42 and Rac1 in ketotic cows. J Dairy Sci 2023; 106:8005-8016. [PMID: 37641273 DOI: 10.3168/jds.2023-23293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/05/2023] [Indexed: 08/31/2023]
Abstract
Dairy cows have high incidence of ketosis during perinatal. According to our previous studies, elevated ketone bodies (mainly β-hydroxybutyrate, BHB) in the peripheral blood are believed to contribute to the impairment of neutrophils mobility and directionality thereby contributing to the immunosuppression and further infectious disease secondary to ketosis. However, the specific effect of BHB on the directionality of bovine neutrophils needs further study and the underlying molecular mechanisms are still unclear. According to the concentration of serum BHB, 40 multiparous cows (within 3 wk postpartum) were selected and divided into the control (n = 20, BHB <0.6 mM) or clinical ketosis (n = 20, BHB >3.0 mM) group. Blood samples were collected for baseline serum characteristics analysis and neutrophil mobility and directionality detection. Platelet activation factor was used as a chemoattractant in cell migration experiments. Our ex-vivo data showed ketotic cows, compared with control cows, were in a negative energy balance state, and their neutrophils had shorter migration distance, lower migration speed, and impaired migration directionality. Neutrophils from control cows were incubated with 3.0 mM BHB for 6 h in vitro. Similarly, BHB stimulation resulted in impaired mobility and directionality of bovine neutrophils. We further specifically studied the underlying molecular mechanism of BHB regulating neutrophil migration directionality in the present study. Cell division control protein 42 homolog (Cdc42) and Ras-related C3 botulinum toxin substrate 1 (Rac1), 2 key markers in the regulation of migration directionality, were found increased after BHB treatment in their total and activated protein levels while decreasing in their transcription level, suggesting that an imbalance of the protein degradation system may be involved. Interestingly, transmission electron microscopy data revealed a decrease in autophagosome number in neutrophils from ketotic cows. Western blotting data showed the accumulation of sequestosome-1 (p62) protein and a decrease in microtubule-associated protein 1 light chain 3-II (LC3-II) protein abundance after BHB treatment, further confirming that the autophagy flux was inhibited in neutrophils from ketotic cows. Additionally, rapamycin (RAPA), a specific autophagy activator, was used with or without BHB treatment in vitro. Accordingly, the BHB-induced impairment of migration directionality but not mobility was relieved by RAPA. Furthermore, as verified by in vivo experiments, compared with the control cows, the protein abundance of total and activated Cdc42 and Rac1 increased and their mRNA abundance decreased in neutrophils from ketotic cows. Overall, the present study revealed that pathological concentration of BHB impairs neutrophil migration directionality through inhibiting the autophagy-mediated degradation of Cdc42 and Rac1. These findings help explain the immunosuppression caused by ketosis.
Collapse
Affiliation(s)
- Yuchen Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shang Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiancheng Feng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chao Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kexin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhe Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
5
|
Qin B, Yu S, Chen Q, Jin LH. Atg2 Regulates Cellular and Humoral Immunity in Drosophila. INSECTS 2023; 14:706. [PMID: 37623416 PMCID: PMC10455222 DOI: 10.3390/insects14080706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
Autophagy is a process that promotes the lysosomal degradation of cytoplasmic proteins and is highly conserved in eukaryotic organisms. Autophagy maintains homeostasis in organisms and regulates multiple developmental processes, and autophagy disruption is related to human diseases. However, the functional roles of autophagy in mediating innate immune responses are largely unknown. In this study, we sought to understand how Atg2, an autophagy-related gene, functions in the innate immunity of Drosophila melanogaster. The results showed that a large number of melanotic nodules were produced upon inhibition of Atg2. In addition, inhibiting Atg2 suppressed the phagocytosis of latex beads, Staphylococcus aureus and Escherichia coli; the proportion of Nimrod C1 (one of the phagocytosis receptors)-positive hemocytes also decreased. Moreover, inhibiting Atg2 altered actin cytoskeleton patterns, showing longer filopodia but with decreased numbers of filopodia. The expression of AMP-encoding genes was altered by inhibiting Atg2. Drosomycin was upregulated, and the transcript levels of Attacin-A, Diptericin and Metchnikowin were decreased. Finally, the above alterations caused by the inhibition of Atg2 prevented flies from resisting invading pathogens, showing that flies with low expression of Atg2 were highly susceptible to Staphylococcus aureus and Erwinia carotovora carotovora 15 infections. In conclusion, Atg2 regulated both cellular and humoral innate immunity in Drosophila. We have identified Atg2 as a crucial regulator in mediating the homeostasis of immunity, which further established the interactions between autophagy and innate immunity.
Collapse
Affiliation(s)
| | | | | | - Li Hua Jin
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.Q.); (S.Y.); (Q.C.)
| |
Collapse
|
6
|
Xing S, Zheng X, Yan H, Mo Y, Duan R, Chen Z, Wang K, Gao K, Chen T, Zhao S, Wang J, Chen L. Superresolution live-cell imaging reveals that the localization of TMEM106B to filopodia in oligodendrocytes is compromised by the hypomyelination-related D252N mutation. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1858-1868. [PMID: 37129766 DOI: 10.1007/s11427-022-2290-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/30/2023] [Indexed: 05/03/2023]
Abstract
Hypomyelination leukodystrophies constitute a group of heritable white matter disorders exhibiting defective myelin development. Initially identified as a lysosomal protein, the TMEM106B D252N mutant has recently been associated with hypomyelination. However, how lysosomal TMEM106B facilitates myelination and how the D252N mutation disrupts that process are poorly understood. We used superresolution Hessian structured illumination microscopy (Hessian-SIM) and spinning disc-confocal structured illumination microscopy (SD-SIM) to find that the wild-type TMEM106B protein is targeted to the plasma membrane, filopodia, and lysosomes in human oligodendrocytes. The D252N mutation reduces the size of lysosomes in oligodendrocytes and compromises lysosome changes upon starvation stress. Most importantly, we detected reductions in the length and number of filopodia in cells expressing the D252N mutant. PLP1 is the most abundant myelin protein that almost entirely colocalizes with TMEM106B, and coexpressing PLP1 with the D252N mutant readily rescues the lysosome and filopodia phenotypes of cells. Therefore, interactions between TMEM106B and PLP1 on the plasma membrane are essential for filopodia formation and myelination in oligodendrocytes, which may be sustained by the delivery of these proteins from lysosomes via exocytosis.
Collapse
Affiliation(s)
- Shijia Xing
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Xiaolu Zheng
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yanquan Mo
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Ruoyu Duan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
- National Center for Children's Health, Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Zhixing Chen
- National Biomedical Imaging Center, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Kunhao Wang
- Key Laboratory of Laser Life Science, Ministry of Education, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Tongsheng Chen
- Key Laboratory of Laser Life Science, Ministry of Education, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Shiqun Zhao
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| | - Liangyi Chen
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
| |
Collapse
|
7
|
Chemoprevention of 4NQO-Induced Mouse Tongue Carcinogenesis by AKT Inhibitor through the MMP-9/RhoC Signaling Pathway and Autophagy. Anal Cell Pathol (Amst) 2022; 2022:3770715. [PMID: 36247874 PMCID: PMC9556259 DOI: 10.1155/2022/3770715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/18/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Oral cancer (OC), the most common cancer in the head and neck, which has a poor prognosis, histopathologically follows a stepwise pattern of hyperplasia, dysplasia, and cancer. Blocking the progression of OC in the precancer stage could greatly improve the survival and cure rates. AKT protein plays a critical role in the signal transduction of cancer cells, and we found that AKT was overexpressed in human OC samples through analysis of TCGA database. Therefore, this study is aimed at investigating the chemopreventive effect of an AKT inhibitor (MK2206 2HCl) on OC. In vivo, we established a 4-nitroquinoline-1-oxide- (4NQO-) induced mouse tongue carcinogenesis model to investigate the potential chemopreventive effect of MK2206 2HCl on mouse OC resulting from 4NQO. The results showed that MK2206 2HCl could significantly reduce the incidence rate and growth of OC, inhibit the transformation of dysplasia to cancer in the 4NQO-induced mouse tongue carcinogenesis model, and simultaneously markedly suppress cell proliferation, angiogenesis, and mast cell (MC) infiltration in 4NQO-induced mouse tongue cancers. In vitro, our results revealed that MK2206 2HCl could also inhibit oral squamous cell carcinoma (OSCC) cell malignant biological behaviors, including cell proliferation, colony formation, cell invasion, and migration, while promoting apoptosis. Mechanistic studies revealed that MK2206 2HCl suppressed matrix metalloproteinase 9 (MMP-9) and RhoC expression and promoted autophagy gene LC3 II expression. In summary, our findings demonstrated the chemopreventive effect of MK2206 2HCl on the 4NQO-induced mouse tongue carcinogenesis model, which likely has an underlying mechanism mediated by the MMP-9/RhoC signaling pathway and autophagy.
Collapse
|
8
|
Molina E, Cataldo VF, Eggers C, Muñoz-Madrid V, Glavic Á. p53 Related Protein Kinase is Required for Arp2/3-Dependent Actin Dynamics of Hemocytes in Drosophila melanogaster. Front Cell Dev Biol 2022; 10:859105. [PMID: 35721516 PMCID: PMC9201722 DOI: 10.3389/fcell.2022.859105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Cells extend membrane protrusions like lamellipodia and filopodia from the leading edge to sense, to move and to form new contacts. The Arp2/3 complex sustains lamellipodia formation, and in conjunction with the actomyosin contractile system, provides mechanical strength to the cell. Drosophila p53-related protein kinase (Prpk), a Tsc5p ortholog, has been described as essential for cell growth and proliferation. In addition, Prpk interacts with proteins associated to actin filament dynamics such as α-spectrin and the Arp2/3 complex subunit Arpc4. Here, we investigated the role of Prpk in cell shape changes, specifically regarding actin filament dynamics and membrane protrusion formation. We found that reductions in Prpk alter cell shape and the structure of lamellipodia, mimicking the phenotypes evoked by Arp2/3 complex deficiencies. Prpk co-localize and co-immunoprecipitates with the Arp2/3 complex subunit Arpc1 and with the small GTPase Rab35. Importantly, expression of Rab35, known by its ability to recruit upstream regulators of the Arp2/3 complex, could rescue the Prpk knockdown phenotypes. Finally, we evaluated the requirement of Prpk in different developmental contexts, where it was shown to be essential for correct Arp2/3 complex distribution and actin dynamics required for hemocytes migration, recruitment, and phagocytosis during immune response.
Collapse
Affiliation(s)
- Emiliano Molina
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Vicente F. Cataldo
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Eggers
- Department for Chemistry and Biochemistry and Pharmaceutical Sciences, Faculty of Science, University of Bern, Bern, Switzerland
| | - Valentina Muñoz-Madrid
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Álvaro Glavic
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- *Correspondence: Álvaro Glavic,
| |
Collapse
|
9
|
Campisi D, Desrues L, Dembélé KP, Mutel A, Parment R, Gandolfo P, Castel H, Morin F. The core autophagy protein ATG9A controls dynamics of cell protrusions and directed migration. J Cell Biol 2022; 221:e202106014. [PMID: 35180289 PMCID: PMC8932524 DOI: 10.1083/jcb.202106014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 01/18/2023] Open
Abstract
Chemotactic migration is a fundamental cellular behavior relying on the coordinated flux of lipids and cargo proteins toward the leading edge. We found here that the core autophagy protein ATG9A plays a critical role in the chemotactic migration of several human cell lines, including highly invasive glioma cells. Depletion of ATG9A protein altered the formation of large and persistent filamentous actin (F-actin)-rich lamellipodia that normally drive directional migration. Using live-cell TIRF microscopy, we demonstrated that ATG9A-positive vesicles are targeted toward the migration front of polarized cells, where their exocytosis correlates with protrusive activity. Finally, we found that ATG9A was critical for efficient delivery of β1 integrin to the leading edge and normal adhesion dynamics. Collectively, our data uncover a new function for ATG9A protein and indicate that ATG9A-positive vesicles are mobilized during chemotactic stimulation to facilitate expansion of the lamellipodium and its anchorage to the extracellular matrix.
Collapse
Affiliation(s)
- Daniele Campisi
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Laurence Desrues
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Kléouforo-Paul Dembélé
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Alexandre Mutel
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Renaud Parment
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Pierrick Gandolfo
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Hélène Castel
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Fabrice Morin
- Normandie University, UNIROUEN, Institut national de la santé et de la recherche médicale U1239, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine, Rouen, France
| |
Collapse
|
10
|
Han NR, Moon PD, Nam SY, Ko SG, Park HJ, Kim HM, Jeong HJ. TSLP up-regulates inflammatory responses through induction of autophagy in T cells. FASEB J 2022; 36:e22148. [PMID: 34997949 DOI: 10.1096/fj.202101447r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022]
Abstract
Thymic stromal lymphopoietin (TSLP), a type I cytokine belonging to the IL-2 cytokine family, promotes Th2-mediated inflammatory responses. The aim of this study is to investigate whether TSLP increases inflammatory responses via induction of autophagy using a murine T cell lymphoma cell line, EL4 cells, and lipopolysaccharide (LPS)-injected mice. TSLP increased expression levels of autophagy-related factors, such as Beclin-1, LC3-II, p62, Atg5, and lysosome associated membrane protein 1/2, whereas these factors increased by TSLP disappeared by neutralization of TSLP in EL4 cells. TSLP activated JAK1/JAK2/STAT5/JNK/PI3K, while the blockade of JAK1/JAK2/STAT5/JNK/PI3K signaling pathways reduced the expression levels of Beclin-1, LC3-II, and p62 in TSLP-stimulated EL4 cells. In addition, TSLP simultaneously increased levels of inflammatory cytokines via induction of autophagy by activation of JAK1/JAK2/STAT5/JNK/PI3K signaling pathways. In an LPS-induced acute liver injury (ALI) mouse model, exogenous TSLP increased expression levels of Beclin-1 and LC3-II, whereas functional deficiency of TSLP by TSLP siRNA resulted in lower expression of Beclin-1, LC3-II, and inflammatory cytokines, impairing their ability to form autophagosomes in ALI mice. Thus, our findings show a new role of TSLP between autophagy and inflammatory responses. In conclusion, regulating TSLP-induced autophagy may be a potential therapeutic strategy for inflammatory responses.
Collapse
Affiliation(s)
- Na-Ra Han
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Phil-Dong Moon
- Center for Converging Humanities, Kyung Hee University, Seoul, Republic of Korea
| | - Sun-Young Nam
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hi-Joon Park
- Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Ja Jeong
- Department of Food Science & Technology, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
11
|
Liu M, Yang J, Xu B, Zhang X. Tumor metastasis: Mechanistic insights and therapeutic interventions. MedComm (Beijing) 2021; 2:587-617. [PMID: 34977870 PMCID: PMC8706758 DOI: 10.1002/mco2.100] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer metastasis is responsible for the vast majority of cancer-related deaths worldwide. In contrast to numerous discoveries that reveal the detailed mechanisms leading to the formation of the primary tumor, the biological underpinnings of the metastatic disease remain poorly understood. Cancer metastasis is a complex process in which cancer cells escape from the primary tumor, settle, and grow at other parts of the body. Epithelial-mesenchymal transition and anoikis resistance of tumor cells are the main forces to promote metastasis, and multiple components in the tumor microenvironment and their complicated crosstalk with cancer cells are closely involved in distant metastasis. In addition to the three cornerstones of tumor treatment, surgery, chemotherapy, and radiotherapy, novel treatment approaches including targeted therapy and immunotherapy have been established in patients with metastatic cancer. Although the cancer survival rate has been greatly improved over the years, it is still far from satisfactory. In this review, we provided an overview of the metastasis process, summarized the cellular and molecular mechanisms involved in the dissemination and distant metastasis of cancer cells, and reviewed the important advances in interventions for cancer metastasis.
Collapse
Affiliation(s)
- Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jing Yang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Bushu Xu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
12
|
Tsai CR, Wang Y, Jacobson A, Sankoorikkal N, Chirinos JD, Burra S, Makthal N, Kumaraswami M, Galko MJ. Pvr and distinct downstream signaling factors are required for hemocyte spreading and epidermal wound closure at Drosophila larval wound sites. G3-GENES GENOMES GENETICS 2021; 12:6423993. [PMID: 34751396 PMCID: PMC8728012 DOI: 10.1093/g3journal/jkab388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022]
Abstract
Tissue injury is typically accompanied by inflammation. In Drosophila melanogaster larvae, wound-induced inflammation involves adhesive capture of hemocytes at the wound surface followed by hemocyte spreading to assume a flat, lamellar morphology. The factors that mediate this cell spreading at the wound site are not known. Here, we discover a role for the platelet-derived growth factor/vascular endothelial growth factor-related receptor (Pvr) and its ligand, Pvf1, in blood cell spreading at the wound site. Pvr and Pvf1 are required for spreading in vivo and in an in vitro spreading assay where spreading can be directly induced by Pvf1 application or by constitutive Pvr activation. In an effort to identify factors that act downstream of Pvr, we performed a genetic screen in which select candidates were tested to determine if they could suppress the lethality of Pvr overexpression in the larval epidermis. Some of the suppressors identified are required for epidermal wound closure (WC), another Pvr-mediated wound response, some are required for hemocyte spreading in vitro, and some are required for both. One of the downstream factors, Mask, is also required for efficient wound-induced hemocyte spreading in vivo. Our data reveal that Pvr signaling is required for wound responses in hemocytes (cell spreading) and defines distinct downstream signaling factors that are required for either epidermal WC or hemocyte spreading.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, United States.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Yan Wang
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Alec Jacobson
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Niki Sankoorikkal
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Josue D Chirinos
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Sirisha Burra
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Nishanth Makthal
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Muthiah Kumaraswami
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Michael J Galko
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, United States.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Genetics & Epigenetics Graduate Program, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
13
|
Semwal MK, Jones NE, Griffith AV. Metabolic Regulation of Thymic Epithelial Cell Function. Front Immunol 2021; 12:636072. [PMID: 33746975 PMCID: PMC7968369 DOI: 10.3389/fimmu.2021.636072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022] Open
Abstract
The thymus is the primary site of T lymphocyte development, where mutually inductive signaling between lymphoid progenitors and thymic stromal cells directs the progenitors along a well-characterized program of differentiation. Although thymic stromal cells, including thymic epithelial cells (TECs) are critical for the development of T cell-mediated immunity, many aspects of their basic biology have been difficult to resolve because they represent a small fraction of thymus cellularity, and because their isolation requires enzymatic digestion that induces broad physiological changes. These obstacles are especially relevant to the study of metabolic regulation of cell function, since isolation procedures necessarily disrupt metabolic homeostasis. In contrast to the well-characterized relationships between metabolism and intracellular signaling in T cell function during an immune response, metabolic regulation of thymic stromal cell function represents an emerging area of study. Here, we review recent advances in three distinct, but interconnected areas: regulation of mTOR signaling, reactive oxygen species (ROS), and autophagy, with respect to their roles in the establishment and maintenance of the thymic stromal microenvironment.
Collapse
Affiliation(s)
- Manpreet K Semwal
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Nicholas E Jones
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Ann V Griffith
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
14
|
Brambilla L, Lahiri T, Cammer M, Levy DE. STAT3 Inhibitor OPB-51602 Is Cytotoxic to Tumor Cells Through Inhibition of Complex I and ROS Induction. iScience 2020; 23:101822. [PMID: 33305182 PMCID: PMC7708861 DOI: 10.1016/j.isci.2020.101822] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/25/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
STAT3 is a transcription factor involved in several cellular activities including inflammation, proliferation, and survival, but it also plays a non-transcriptional role in modulating mitochondrial metabolism. Given its diverse functions in human cancers, it is an emerging therapeutic target. Here we show that OPB-51602, a small molecule inhibitor of STAT3, is highly toxic in a STAT3-dependent manner. Specifically, drug toxicity depends on mitochondrial STAT3 as tumor cells expressing only a mitochondrially restricted form of STAT3 are sensitive to the compound, whereas STAT3-null cells are protected. OPB-51602 inhibited complex I activity and led to increased ROS production, which in turn induced mitophagy, actin rearrangements, and cell death. Cells undergoing reduced oxidative phosphorylation or expressing NDI1 NADH dehydrogenase from Saccharomyces cerevisiae, which bypasses mammalian complex I, were resistant to OPB-51602 toxicity. These results show that targeting mitochondrial STAT3 function causes synthetic lethality through complex I inhibition that could be exploited for cancer chemotherapy. OPB-51602 is cytotoxic to human tumor cell lines in a STAT3-dependent manner Cytotoxicity depends on ROS induction and leads to mitophagy and actin remodeling OPB-51602 affects oxidative phosphorylation by inhibiting complex I via STAT3 Expression of a STAT3-independent form of complex I is cytoprotective
Collapse
Affiliation(s)
- Lara Brambilla
- Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, 550 1st Avenue MSB548A, New York, NY 10016, USA
| | - Tanaya Lahiri
- Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, 550 1st Avenue MSB548A, New York, NY 10016, USA
| | - Michael Cammer
- Microscopy Core, Division of Advanced Research Technologies, NYU Grossman School of Medicine, 55- 1st Avenue SK2, New York, NY 10016, USA
| | - David E Levy
- Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, 550 1st Avenue MSB548A, New York, NY 10016, USA
| |
Collapse
|
15
|
Vasilevich AS, Vermeulen S, Kamphuis M, Roumans N, Eroumé S, Hebels DGAJ, van de Peppel J, Reihs R, Beijer NRM, Carlier A, Carpenter AE, Singh S, de Boer J. On the correlation between material-induced cell shape and phenotypical response of human mesenchymal stem cells. Sci Rep 2020; 10:18988. [PMID: 33149200 PMCID: PMC7642380 DOI: 10.1038/s41598-020-76019-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
Learning rules by which cell shape impacts cell function would enable control of cell physiology and fate in medical applications, particularly, on the interface of cells and material of the implants. We defined the phenotypic response of human bone marrow-derived mesenchymal stem cells (hMSCs) to 2176 randomly generated surface topographies by probing basic functions such as migration, proliferation, protein synthesis, apoptosis, and differentiation using quantitative image analysis. Clustering the surfaces into 28 archetypical cell shapes, we found a very strict correlation between cell shape and physiological response and selected seven cell shapes to describe the molecular mechanism leading to phenotypic diversity. Transcriptomics analysis revealed a tight link between cell shape, molecular signatures, and phenotype. For instance, proliferation is strongly reduced in cells with limited spreading, resulting in down-regulation of genes involved in the G2/M cycle and subsequent quiescence, whereas cells with large filopodia are related to activation of early response genes and inhibition of the osteogenic process. In this paper we were aiming to identify a universal set of genes that regulate the material-induced phenotypical response of human mesenchymal stem cells. This will allow designing implants that can actively regulate cellular, molecular signalling through cell shape. Here we are proposing an approach to tackle this question.
Collapse
Affiliation(s)
- Aliaksei S Vasilevich
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Steven Vermeulen
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marloes Kamphuis
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nadia Roumans
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Said Eroumé
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dennie G A J Hebels
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rika Reihs
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nick R M Beijer
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jan de Boer
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
16
|
Kang R, Zeh H, Lotze M, Tang D. The Multifaceted Effects of Autophagy on the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:99-114. [PMID: 32030650 DOI: 10.1007/978-3-030-35727-6_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is composed of cancer cells, noncancer cells (e.g., immune cells, stromal cells, endothelial cells, and adipocytes), and various mediators (e.g., cytokines, chemokines, growth factors, and humoral factors) that work together to support cancer growth, progression, and resistance to therapies. Autophagy is an evolutionarily conserved degradation mechanism by which various cytosolic cargos (e.g., damaged organelles, unused molecules, or invaded pathogens) are engulfed by double-membrane autophagosomes, and then delivered into the lysosome for degradation and recycling. The level of autophagy is a crucial threshold to either promote cell survival or induce cell death in response to environmental stresses. Autophagy plays a context-dependent role in tumorigenesis and anticancer therapy via shaping the inflammatory, hypoxic, immunosuppressive, and metabolic tumor microenvironment. In particular, impaired autophagy flux is associated with chronic inflammation, immunosuppression, stromal formation, cancer stemness, angiogenesis, metastasis, and metabolic reprogramming in the tumor microenvironment. Understanding the molecular machinery of autophagy and its communication with hallmarks of cancer could lead to potential new anticancer strategies or drugs.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Impaired Autophagy in the Fibroblasts by Titanium Particles Increased the Release of CX3CL1 and Promoted the Chemotactic Migration of Monocytes. Inflammation 2019; 43:673-685. [DOI: 10.1007/s10753-019-01149-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Bankston AN, Forston MD, Howard RM, Andres KR, Smith AE, Ohri SS, Bates ML, Bunge MB, Whittemore SR. Autophagy is essential for oligodendrocyte differentiation, survival, and proper myelination. Glia 2019; 67:1745-1759. [PMID: 31162728 DOI: 10.1002/glia.23646] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/01/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Deficient myelination, the spiral wrapping of highly specialized membrane around axons, causes severe neurological disorders. Maturation of oligodendrocyte progenitor cells (OPC) to myelinating oligodendrocytes (OL), the sole providers of central nervous system (CNS) myelin, is tightly regulated and involves extensive morphological changes. Here, we present evidence that autophagy, the targeted isolation of cytoplasm and organelles by the double-membrane autophagosome for lysosomal degradation, is essential for OPC/OL differentiation, survival, and proper myelin development. A marked increase in autophagic activity coincides with OL differentiation, with OL processes having the greatest increase in autophagic flux. Multiple lines of evidence indicate that autophagosomes form in developing myelin sheathes before trafficking from myelin to the OL soma. Mice with conditional OPC/OL-specific deletion of the essential autophagy gene Atg5 beginning on postnatal Day 5 develop a rapid tremor and die around postnatal Day 12. Further analysis revealed apoptotic death of OPCs, reduced differentiation, and reduced myelination. Surviving Atg5-/- OLs failed to produce proper myelin structure. In vitro, pharmacological inhibition of autophagy in OPC/dorsal root ganglion (DRG) co-cultures blocked myelination, producing OLs surrounded by many short processes. Conversely, autophagy stimulation enhanced myelination. These results implicate autophagy as a key regulator of OPC survival, maturation, and proper myelination. Autophagy may provide an attractive target to promote both OL survival and subsequent myelin repair after injury.
Collapse
Affiliation(s)
- Andrew N Bankston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Michael D Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Allison E Smith
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Margaret L Bates
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Mary B Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida.,Department of Cell Biology and Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
19
|
Fontana R, Ranieri M, La Mantia G, Vivo M. Dual Role of the Alternative Reading Frame ARF Protein in Cancer. Biomolecules 2019; 9:E87. [PMID: 30836703 PMCID: PMC6468759 DOI: 10.3390/biom9030087] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
The CDKN2a/ARF locus expresses two partially overlapping transcripts that encode two distinct proteins, namely p14ARF (p19Arf in mouse) and p16INK4a, which present no sequence identity. Initial data obtained in mice showed that both proteins are potent tumor suppressors. In line with a tumor-suppressive role, ARF-deficient mice develop lymphomas, sarcomas, and adenocarcinomas, with a median survival rate of one year of age. In humans, the importance of ARF inactivation in cancer is less clear whereas a more obvious role has been documented for p16INK4a. Indeed, many alterations in human tumors result in the elimination of the entire locus, while the majority of point mutations affect p16INK4a. Nevertheless, specific mutations of p14ARF have been described in different types of human cancers such as colorectal and gastric carcinomas, melanoma and glioblastoma. The activity of the tumor suppressor ARF has been shown to rely on both p53-dependent and independent functions. However, novel data collected in the last years has challenged the traditional and established role of this protein as a tumor suppressor. In particular, tumors retaining ARF expression evolve to metastatic and invasive phenotypes and in humans are associated with a poor prognosis. In this review, the recent evidence and the molecular mechanisms of a novel role played by ARF will be presented and discussed, both in pathological and physiological contexts.
Collapse
Affiliation(s)
- Rosa Fontana
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Michela Ranieri
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY 10016, USA.
| | - Girolama La Mantia
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Maria Vivo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| |
Collapse
|
20
|
Hao Y, Yu S, Luo F, Jin LH. Jumu is required for circulating hemocyte differentiation and phagocytosis in Drosophila. Cell Commun Signal 2018; 16:95. [PMID: 30518379 PMCID: PMC6280549 DOI: 10.1186/s12964-018-0305-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/19/2018] [Indexed: 11/15/2022] Open
Abstract
Background The regulatory mechanisms of hematopoiesis and cellular immunity show a high degree of similarity between insects and mammals, and Drosophila has become a good model for investigating cellular immune responses. Jumeau (Jumu) is a member of the winged-helix/forkhead (FKH) transcription factor family and is required for Drosophila development. Adult jumu mutant flies show defective hemocyte phagocytosis and a weaker defense capability against pathogen infection. Here, we further investigated the role of jumu in the regulation of larval hemocyte development and phagocytosis. Methods In vivo phagocytosis assays, immunohistochemistry, Real-time quantitative PCR and immunoblotting were performed to investigate the effect of Jumu on hemocyte phagocytosis. 5-Bromo-2-deoxyUridine (BrdU) labeling, phospho-histone H3 (PH3) and TdT-mediated dUTP Nick-End Labeling (TUNEL) staining were performed to analyze the proliferation and apoptosis of hemocyte; immunohistochemistry and Mosaic analysis with a repressible cell marker (MARCM) clone analysis were performed to investigate the role of Jumu in the activation of Toll pathway. Results Jumu indirectly controls hemocyte phagocytosis by regulating the expression of NimC1 and cytoskeleton reorganization. The loss of jumu also causes abnormal proliferation and differentiation in circulating hemocytes. Our results suggest that a severe deficiency of jumu leads to the generation of enlarged multinucleate hemocytes by affecting the normal cell mitosis process and induces numerous lamellocytes by activating the Toll pathway. Conclusions Jumu regulates circulating hemocyte differentiation and phagocytosis in Drosophila. Our findings provide new insight into the mechanistic roles of cytoskeleton regulatory proteins in phagocytosis and establish a basis for further analyses of the regulatory mechanism of the mammalian ortholog of Jumu in mammalian innate immunity. Electronic supplementary material The online version of this article (10.1186/s12964-018-0305-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yangguang Hao
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, People's Republic of China.,Department of Translational medicine research center, Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Shichao Yu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, People's Republic of China
| | - Fangzhou Luo
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, People's Republic of China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, People's Republic of China.
| |
Collapse
|
21
|
Clark SG, Graybeal LL, Bhattacharjee S, Thomas C, Bhattacharya S, Cox DN. Basal autophagy is required for promoting dendritic terminal branching in Drosophila sensory neurons. PLoS One 2018; 13:e0206743. [PMID: 30395636 PMCID: PMC6218061 DOI: 10.1371/journal.pone.0206743] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/18/2018] [Indexed: 01/22/2023] Open
Abstract
Dendrites function as the primary sites for synaptic input and integration with impairments in dendritic arborization being associated with dysfunctional neuronal circuitry. Post-mitotic neurons require high levels of basal autophagy to clear cytotoxic materials and autophagic dysfunction under native or cellular stress conditions has been linked to neuronal cell death as well as axo-dendritic degeneration. However, relatively little is known regarding the developmental role of basal autophagy in directing aspects of dendritic arborization or the mechanisms by which the autophagic machinery may be transcriptionally regulated to promote dendritic diversification. We demonstrate that autophagy-related (Atg) genes are positively regulated by the homeodomain transcription factor Cut, and that basal autophagy functions as a downstream effector pathway for Cut-mediated dendritic terminal branching in Drosophila multidendritic (md) sensory neurons. Further, loss of function analyses implicate Atg genes in promoting cell type-specific dendritic arborization and terminal branching, while gain of function studies suggest that excessive autophagy leads to dramatic reductions in dendritic complexity. We demonstrate that the Atg1 initiator kinase interacts with the dual leucine zipper kinase (DLK) pathway by negatively regulating the E3 ubiquitin ligase Highwire and positively regulating the MAPKKK Wallenda. Finally, autophagic induction partially rescues dendritic atrophy defects observed in a model of polyglutamine toxicity. Collectively, these studies implicate transcriptional control of basal autophagy in directing dendritic terminal branching and demonstrate the importance of homeostatic control of autophagic levels for dendritic arbor complexity under native or cellular stress conditions.
Collapse
Affiliation(s)
- Sarah G. Clark
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Lacey L. Graybeal
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Shatabdi Bhattacharjee
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Caroline Thomas
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Surajit Bhattacharya
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
22
|
Li K, Zhang JH, Yang YJ, Han W, Yin H. Morphology and fine organization of the midgut of Gampsocleis gratiosa (Orthoptera: Tettigoniidae). PLoS One 2018; 13:e0200405. [PMID: 29985965 PMCID: PMC6037380 DOI: 10.1371/journal.pone.0200405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/26/2018] [Indexed: 11/18/2022] Open
Abstract
The morphology and ultrastructure of the midgut of Gampsocleis gratiosa (Orthoptera: Tettigoniidae) was examined by light and electron microscopy. The midgut consists of two bulbous gastric caeca and a tubular ventriculus. The general organization of the gastric caeca is similar to that of the ventriculus. They are composed of a peritrophic membrane, an epithelium, a basal lamina and muscle layer from the inside to outside. Three types of cells were identified: regenerative, principal, and endocrine. Regenerative cells occur in groups (called nidi) at the base of principal cells. Principal cells grow from regenerative cells. Rare endocrine cells are scattered throughout the epithelium. Principal cells exhibit intense secretory activity, and regional differences in their ultrastructure were observed along the entire midgut. The microvilli are longer than those in any other region in the posterior region of the midgut. Lysosomes, multivesicular bodies (MVBs), autophagosomes, abundant Golgi apparatuses and lipid droplets primarily occur in the gastric caeca. Three pathways of secretion (merocrine, apocrine and holocrine) occur within the midgut epithelium, and a distinctive type of apocrine bleb was found in the gastric caeca. Therefore, these gastric caeca may be evolving toward a special type of gland.
Collapse
Affiliation(s)
- Ke Li
- College of Life Sciences, Shanxi Normal University, Linfen, Shanxi, China
- Modern College of Humanities and Sciences, Shanxi Normal University, Linfen, Shanxi, China
| | - Jin-Hui Zhang
- College of Life Sciences, Shanxi Normal University, Linfen, Shanxi, China
| | - Yu-Jing Yang
- College of Life Sciences, Shanxi Normal University, Linfen, Shanxi, China
| | - Wei Han
- Modern College of Humanities and Sciences, Shanxi Normal University, Linfen, Shanxi, China
| | - Huan Yin
- College of Life Sciences, Shanxi Normal University, Linfen, Shanxi, China
- * E-mail:
| |
Collapse
|
23
|
Fontana R, Vivo M. Dynamics of p14ARF and Focal Adhesion Kinase-Mediated Autophagy in Cancer. Cancers (Basel) 2018; 10:cancers10070221. [PMID: 29966311 PMCID: PMC6071150 DOI: 10.3390/cancers10070221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 06/22/2018] [Accepted: 06/26/2018] [Indexed: 12/23/2022] Open
Abstract
It has been widely shown that the focal adhesion kinase (FAK) is involved in nearly every aspect of cancer, from invasion to metastasis to epithelial–mesenchymal transition and maintenance of cancer stem cells. FAK has been shown to interact with p14ARF (alternative reading frame)—a well-established tumor suppressor—and functions in the negative regulation of cancer through both p53-dependent and -independent pathways. Interestingly, both FAK and ARF (human and mouse counterpart) proteins, as well as p53, are involved in autophagy—a process of “self-digestion”—whose main function is the recycling of cellular components and quality control of proteins and organelles. In the last years, an unexpected role of p14ARF in the survival of cancer cells has been underlined in different cellular contexts, suggesting a novel pro-oncogenic function of this protein. In this review, the mechanisms whereby ARF and FAK control autophagy are presented, as well as the role of autophagy in cell migration and spreading. Integrated investigation of these cell functions is extremely important to understand the mechanism of the basis of cell transformation and migration and thus cancer development.
Collapse
Affiliation(s)
- Rosa Fontana
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Maria Vivo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| |
Collapse
|
24
|
Lei J, Calvo P, Vigh R, Burd I. Journey to the Center of the Fetal Brain: Environmental Exposures and Autophagy. Front Cell Neurosci 2018; 12:118. [PMID: 29773977 PMCID: PMC5943497 DOI: 10.3389/fncel.2018.00118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/13/2018] [Indexed: 01/28/2023] Open
Abstract
Fetal brain development is known to be affected by adverse environmental exposures during pregnancy, including infection, inflammation, hypoxia, alcohol, starvation, and toxins. These exposures are thought to alter autophagy activity in the fetal brain, leading to adverse perinatal outcomes, such as cognitive and sensorimotor deficits. This review introduces the physiologic autophagy pathways in the fetal brain. Next, methods to detect and monitor fetal brain autophagy activity are outlined. An additional discussion explores possible mechanisms by which environmental exposures during pregnancy alter fetal brain autophagy activity. In the final section, a correlation of fetal autophagy activity with the observed postnatal phenotype is attempted. Our main purpose is to provide the current understanding or a lack thereof mechanisms on autophagy, underlying the fetal brain injury exposed to environmental insults.
Collapse
Affiliation(s)
- Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pilar Calvo
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard Vigh
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Matsumoto C, Jiang Y, Emathinger J, Quijada P, Nguyen N, De La Torre A, Moshref M, Nguyen J, Levinson AB, Shin M, Sussman MA, Hariharan N. Short Telomeres Induce p53 and Autophagy and Modulate Age-Associated Changes in Cardiac Progenitor Cell Fate. Stem Cells 2018; 36:868-880. [PMID: 29441645 DOI: 10.1002/stem.2793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/07/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Aging severely limits myocardial repair and regeneration. Delineating the impact of age-associated factors such as short telomeres is critical to enhance the regenerative potential of cardiac progenitor cells (CPCs). We hypothesized that short telomeres activate p53 and induce autophagy to elicit the age-associated change in CPC fate. We isolated CPCs and compared mouse strains with different telomere lengths for phenotypic characteristics of aging. Wild mouse strain Mus musculus castaneus (CAST) possessing short telomeres exhibits early cardiac aging with cardiac dysfunction, hypertrophy, fibrosis, and senescence, as compared with common lab strains FVB and C57 bearing longer telomeres. CAST CPCs with short telomeres demonstrate altered cell fate as characterized by cell cycle arrest, senescence, basal commitment, and loss of quiescence. Elongation of telomeres using a modified mRNA for telomerase restores youthful properties to CAST CPCs. Short telomeres induce autophagy in CPCs, a catabolic protein degradation process, as evidenced by reduced p62 and increased accumulation of autophagic puncta. Pharmacological inhibition of autophagosome formation reverses the cell fate to a more youthful phenotype. Mechanistically, cell fate changes induced by short telomeres are partially p53 dependent, as p53 inhibition rescues senescence and commitment observed in CAST CPCs, coincident with attenuation of autophagy. In conclusion, short telomeres activate p53 and autophagy to tip the equilibrium away from quiescence and proliferation toward differentiation and senescence, leading to exhaustion of CPCs. This study provides the mechanistic basis underlying age-associated cell fate changes that will enable identification of molecular strategies to prevent senescence of CPCs. Stem Cells 2018;36:868-880.
Collapse
Affiliation(s)
- Collin Matsumoto
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Yan Jiang
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | | | - Pearl Quijada
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Nathalie Nguyen
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Andrea De La Torre
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Maryam Moshref
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Jonathan Nguyen
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Aimee B Levinson
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Minyoung Shin
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Mark A Sussman
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Nirmala Hariharan
- Department of Pharmacology, University of California at Davis, Davis, California, USA.,Department of Biology, San Diego State University, San Diego, California, USA
| |
Collapse
|
26
|
Mowers EE, Sharifi MN, Macleod KF. Functions of autophagy in the tumor microenvironment and cancer metastasis. FEBS J 2018; 285:1751-1766. [PMID: 29356327 DOI: 10.1111/febs.14388] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/07/2018] [Accepted: 01/16/2018] [Indexed: 02/06/2023]
Abstract
Macro-autophagy is an ancient and highly conserved self-degradative process that plays a homeostatic role in normal cells by eliminating organelles, pathogens, and protein aggregates. Autophagy, as it is routinely referred to, also allows cells to maintain metabolic sufficiency and survive under conditions of nutrient stress by recycling the by-products of autophagic degradation, such as fatty acids, amino acids, and nucleotides. Tumor cells are more reliant than normal cells on autophagy for survival in part due to their rapid growth rate, altered metabolism, and nutrient-deprived growth environment. How this dependence of tumor cells on autophagy affects their progression to malignancy and metastatic disease is an area of increasing research focus. Here, we review recent work identifying critical functions for autophagy in tumor cell migration and invasion, tumor stem cell maintenance and therapy resistance, and cross-talk between tumor cells and their microenvironment.
Collapse
Affiliation(s)
- Erin E Mowers
- The Ben May Department for Cancer Research, University of Chicago, IL, USA.,The Committee on Cancer Biology, Chicago, IL, USA.,Inter-disciplinary Scientist Training Program, Chicago, IL, USA
| | - Marina N Sharifi
- The Ben May Department for Cancer Research, University of Chicago, IL, USA.,The Committee on Cancer Biology, Chicago, IL, USA.,Medical Scientist Training Program, Chicago, IL, USA
| | - Kay F Macleod
- The Ben May Department for Cancer Research, University of Chicago, IL, USA.,The Committee on Cancer Biology, Chicago, IL, USA.,The University of Chicago, IL, USA
| |
Collapse
|
27
|
Sanchez-Vera V, Kenchappa CS, Landberg K, Bressendorff S, Schwarzbach S, Martin T, Mundy J, Petersen M, Thelander M, Sundberg E. Autophagy is required for gamete differentiation in the moss Physcomitrella patens. Autophagy 2017; 13:1939-1951. [PMID: 28837383 PMCID: PMC5788497 DOI: 10.1080/15548627.2017.1366406] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/17/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022] Open
Abstract
Autophagy, a major catabolic process in eukaryotes, was initially related to cell tolerance to nutrient depletion. In plants autophagy has also been widely related to tolerance to biotic and abiotic stresses (through the induction or repression of programmed cell death, PCD) as well as to promotion of developmentally regulated PCD, starch degradation or caloric restriction important for life span. Much less is known regarding its role in plant cell differentiation. Here we show that macroautophagy, the autophagy pathway driven by engulfment of cytoplasmic components by autophagosomes and its subsequent degradation in vacuoles, is highly active during germ cell differentiation in the early diverging land plant Physcomitrella patens. Our data provide evidence that suppression of ATG5-mediated autophagy results in reduced density of the egg cell-mediated mucilage that surrounds the mature egg, pointing toward a potential role of autophagy in extracellular mucilage formation. In addition, we found that ATG5- and ATG7-mediated autophagy is essential for the differentiation and cytoplasmic reduction of the flagellated motile sperm and hence for sperm fertility. The similarities between the need of macroautophagy for sperm differentiation in moss and mouse are striking, strongly pointing toward an ancestral function of autophagy not only as a protector against nutrient stress, but also in gamete differentiation.
Collapse
Affiliation(s)
- Victoria Sanchez-Vera
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | - Chandra Shekar Kenchappa
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | - Katarina Landberg
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | | | - Stefan Schwarzbach
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | - Tom Martin
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | - John Mundy
- Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Morten Petersen
- Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Mattias Thelander
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| | - Eva Sundberg
- Department of Plant Biology, Swedish University of Agricultural Sciences, The Linnean Centre of Plant Biology in Uppsala, Uppsala, Sweden
| |
Collapse
|
28
|
Singh A, Sen E. Reciprocal role of SIRT6 and Hexokinase 2 in the regulation of autophagy driven monocyte differentiation. Exp Cell Res 2017; 360:365-374. [PMID: 28935467 DOI: 10.1016/j.yexcr.2017.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/16/2022]
Abstract
Emerging evidences suggest the impact of autophagy on differentiation but the underlying molecular links between metabolic restructuring and autophagy during monocyte differentiation remain elusive. An increase in PPARγ, HK2 and SIRT6 expression was observed upon PMA induced monocyte differentiation. While PPARγ positively regulated HK2 and SIRT6 expression, the latter served as a negative regulator of HK2. Changes in expression of these metabolic modelers were accompanied by decreased glucose uptake and increase in Chibby, a potent antagonist of β-catenin/Wnt pathway. Knockdown of Chibby abrogated PMA induced differentiation. While inhibition of HK2 either by Lonidamine or siRNA further elevated PMA induced Chibby, mitochondrial ROS, TIGAR and LC3II levels; siRNA mediated knock-down of SIRT6 exhibited contradictory effects as compared to HK2. Notably, inhibition of autophagy increased HK2, diminished Chibby level and CD33 expression. In addition, PMA induced expression of cytoskeletal architectural proteins, CXCR4, phagocytosis, acquisition of macrophage phenotypes and release of pro-inflammatory mediators was found to be HK2 dependent. Collectively, our findings highlight the previously unknown reciprocal influence of SIRT6 and HK2 in regulating autophagy driven monocyte differentiation.
Collapse
Affiliation(s)
- Ankita Singh
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Ellora Sen
- National Brain Research Centre, Manesar, Haryana 122051, India.
| |
Collapse
|
29
|
Autophagy in cancer metastasis. Oncogene 2016; 36:1619-1630. [PMID: 27593926 PMCID: PMC5337449 DOI: 10.1038/onc.2016.333] [Citation(s) in RCA: 358] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 07/25/2016] [Accepted: 07/31/2016] [Indexed: 02/07/2023]
Abstract
Autophagy is a highly conserved self-degradative process that has a key role in cellular stress responses and survival. Recent work has begun to explore the function of autophagy in cancer metastasis, which is of particular interest given the dearth of effective therapeutic options for metastatic disease. Autophagy is induced upon progression of various human cancers to metastasis and together with data from genetically engineered mice and experimental metastasis models, a role for autophagy at nearly every phase of the metastatic cascade has been identified. Specifically, autophagy has been shown to be involved in modulating tumor cell motility and invasion, cancer stem cell viability and differentiation, resistance to anoikis, epithelial-to-mesenchymal transition, tumor cell dormancy and escape from immune surveillance, with emerging functions in establishing the pre-metastatic niche and other aspects of metastasis. In this review, we provide a general overview of how autophagy modulates cancer metastasis and discuss the significance of new findings for disease management.
Collapse
|
30
|
Schmid MR, Anderl I, Vo HTM, Valanne S, Yang H, Kronhamn J, Rämet M, Rusten TE, Hultmark D. Genetic Screen in Drosophila Larvae Links ird1 Function to Toll Signaling in the Fat Body and Hemocyte Motility. PLoS One 2016; 11:e0159473. [PMID: 27467079 PMCID: PMC4965076 DOI: 10.1371/journal.pone.0159473] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/05/2016] [Indexed: 12/26/2022] Open
Abstract
To understand how Toll signaling controls the activation of a cellular immune response in Drosophila blood cells (hemocytes), we carried out a genetic modifier screen, looking for deletions that suppress or enhance the mobilization of sessile hemocytes by the gain-of-function mutation Toll10b (Tl10b). Here we describe the results from chromosome arm 3R, where five regions strongly suppressed this phenotype. We identified the specific genes immune response deficient 1 (ird1), headcase (hdc) and possibly Rab23 as suppressors, and we studied the role of ird1 in more detail. An ird1 null mutant and a mutant that truncates the N-terminal kinase domain of the encoded Ird1 protein affected the Tl10b phenotype, unlike mutations that affect the C-terminal part of the protein. The ird1 null mutant suppressed mobilization of sessile hemocytes, but enhanced other Tl10b hemocyte phenotypes, like the formation of melanotic nodules and the increased number of circulating hemocytes. ird1 mutants also had blood cell phenotypes on their own. They lacked crystal cells and showed aberrant formation of lamellocytes. ird1 mutant plasmatocytes had a reduced ability to spread on an artificial substrate by forming protrusions, which may explain why they did not go into circulation in response to Toll signaling. The effect of the ird1 mutation depended mainly on ird1 expression in hemocytes, but ird1-dependent effects in other tissues may contribute. Specifically, the Toll receptor was translocated from the cell membrane to intracellular vesicles in the fat body of the ird1 mutant, and Toll signaling was activated in that tissue, partially explaining the Tl10b-like phenotype. As ird1 is otherwise known to control vesicular transport, we conclude that the vesicular transport system may be of particular importance during an immune response.
Collapse
Affiliation(s)
| | - Ines Anderl
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- BioMediTech, University of Tampere, Tampere, Finland
| | - Hoa T. M. Vo
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | - Hairu Yang
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jesper Kronhamn
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Mika Rämet
- BioMediTech, University of Tampere, Tampere, Finland
- PEDEGO Research Center, and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tor Erik Rusten
- Department of Molecular Cell Biology, Oslo University Hospital, Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Dan Hultmark
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- BioMediTech, University of Tampere, Tampere, Finland
| |
Collapse
|
31
|
Kumar S, Kim Y. Glyceraldehyde-3-phosphate dehydrogenase is a mediator of hemocyte-spreading behavior and molecular target of immunosuppressive factor CrV1. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 54:97-108. [PMID: 26366678 DOI: 10.1016/j.dci.2015.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/09/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Cellular immunity is accompanied by hemocyte-spreading behavior, which undergoes cytoskeletal rearrangement. Polydnaviral factor CpBV-CrV1 can inhibit the hemocyte-spreading behavior and suppress host immune response of Plutella xylostella. However, host target molecule of CpBV-CrV1 that inhibits the hemocyte behavior has not been identified yet. This study used a pull-down approach to identify the target molecule of CpBV-CrV1. A protein bound to CpBV-CrV1 was co-precipitated and identified to be glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by LC-MS/MS analysis. RNA interference (RNAi) specific to GAPDH of P. xylostella was found to be able to inhibit the hemocyte-spreading behavior, while RNAi treatments with other glycolytic genes had no effect on the spreading behavior. An addition of recombinant CpBV-CrV1 on hemocyte monolayer interrupted the association between GAPDH and α-tubulin in the cytoplasm. Overlay of mutant proteins (Y492A or Y501A with tyrosine to alanine at putative GAPDH-binding site) of CpBV-CrV1 on hemocyte monolayer revealed that they could enter hemocytes unlike a mutant in the N-terminal coiled-coil domain. However, they failed to inhibit the hemocyte-spreading behavior without any binding affinity to GAPDH. These results suggest that GAPDH plays a critical role in hemocyte-spreading behavior during immune challenge as a molecular target of viral factor CpBV-CrV1.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Bioresource Sciences, Andong National University, Andong 760-749, Republic of Korea
| | - Yonggyun Kim
- Department of Bioresource Sciences, Andong National University, Andong 760-749, Republic of Korea.
| |
Collapse
|
32
|
Xu Z, Yang L, Xu S, Zhang Z, Cao Y. The receptor proteins: pivotal roles in selective autophagy. Acta Biochim Biophys Sin (Shanghai) 2015; 47:571-580. [PMID: 26112016 DOI: 10.1093/abbs/gmv055] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/30/2015] [Indexed: 02/05/2023] Open
Abstract
Autophagy is a highly regulated and multistep biological process whereby cells under metabolic, proteotoxic, or other stresses remove dysfunctional organelles and/or misfolded/polyubiquitinated proteins by shuttling them via specialized structures called autophagosomes to the lysosome for degradation. Although autophagy is generally considered to be a non-selective process, accumulating evidence suggests that it can also selectively degrade specific target cargoes. These selective targets include proteins, mitochondria, and even invading bacteria. The discovery and characterization of autophagic adapters, such as p62/Sequestosome 1 (SQSTM1) and Neighbor of BRCA1 gene 1 (NBR1), have provided mechanistic insights into selective autophagy. These receptors are all able to act as cargo receptors for the degradation of ubiquitinated substrates. This review mainly summarizes the most up-to-date findings regarding the key receptor proteins that play important roles in regulating selective autophagy.
Collapse
Affiliation(s)
- Zhijie Xu
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Lifang Yang
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - San Xu
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Zhibao Zhang
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Ya Cao
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| |
Collapse
|
33
|
Stranks AJ, Hansen AL, Panse I, Mortensen M, Ferguson DJP, Puleston DJ, Shenderov K, Watson AS, Veldhoen M, Phadwal K, Cerundolo V, Simon AK. Autophagy Controls Acquisition of Aging Features in Macrophages. J Innate Immun 2015; 7:375-91. [PMID: 25764971 PMCID: PMC4386145 DOI: 10.1159/000370112] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 12/14/2022] Open
Abstract
Macrophages provide a bridge linking innate and adaptive immunity. An increased frequency of macrophages and other myeloid cells paired with excessive cytokine production is commonly seen in the aging immune system, known as 'inflamm-aging'. It is presently unclear how healthy macrophages are maintained throughout life and what connects inflammation with myeloid dysfunction during aging. Autophagy, an intracellular degradation mechanism, has known links with aging and lifespan extension. Here, we show for the first time that autophagy regulates the acquisition of major aging features in macrophages. In the absence of the essential autophagy gene Atg7, macrophage populations are increased and key functions such as phagocytosis and nitrite burst are reduced, while the inflammatory cytokine response is significantly increased - a phenotype also observed in aged macrophages. Furthermore, reduced autophagy decreases surface antigen expression and skews macrophage metabolism toward glycolysis. We show that macrophages from aged mice exhibit significantly reduced autophagic flux compared to young mice. These data demonstrate that autophagy plays a critical role in the maintenance of macrophage homeostasis and function, regulating inflammation and metabolism and thereby preventing immunosenescence. Thus, autophagy modulation may prevent excess inflammation and preserve macrophage function during aging, improving immune responses and reducing the morbidity and mortality associated with inflamm-aging.
Collapse
Affiliation(s)
- Amanda J Stranks
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tang Z, Zhang N, Di W, Li W. Inhibition of microtubule-associated protein 1 light chain 3B via small-interfering RNA or 3-methyladenine impairs hypoxia-induced HO8910PM and HO8910 epithelial ovarian cancer cell migration and invasion and is associated with RhoA and alterations of the actin cytoskeleton. Oncol Rep 2015; 33:1411-7. [PMID: 25607473 DOI: 10.3892/or.2015.3742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/22/2014] [Indexed: 11/06/2022] Open
Abstract
Expression of microtubule‑associated protein 1 light chain 3 (LC3) is correlated with poor prognosis in many human cancers. Hypoxia induces LC3 expression and is an essential characteristic of epithelial ovarian cancer (EOC). The aim of the present study was to elucidate the mechanism by which LC3 facilitates EOC cell migration and invasion under conditions of hypoxia. The effects of LC3B inhibition under hypoxic conditions on migration, invasion, and adhesion in HO8910PM and HO8910 EOC cell lines were investigated. LC3B inhibition was achieved by small‑interfering RNA (siRNA) targeting LC3B or by treatment with 3‑methyladenine (3‑MA). Cell migration, invasion and adhesion and the arrangement of the cytoskeleton were determined by Transwell migration assays and rhodamine phalloidin staining. Western blot analysis was performed to evaluate the expression level of LC3B and the expression and activity of ras homolog gene family member A (RhoA). Increased LC3B expression was associated with HO8910PM and HO8910 cell migration and invasion promoted under hypoxic conditions. LC3B siRNA and 3‑MA treatment each attenuated hypoxia‑induced LC3B expression, along with migration and invasion, and this was associated with a decrease in RhoA expression and disorganization of the actin cytoskeleton. LC3B may promote the migration and invasion of EOC cells by affecting the cytoskeleton via the RhoA pathway. In addition, LC3B may be a marker of tumor hypoxia and/or metastasis in EOC cells.
Collapse
Affiliation(s)
- Zhongyuan Tang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ning Zhang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Weiping Li
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
35
|
How and why to study autophagy in Drosophila: it's more than just a garbage chute. Methods 2014; 75:151-61. [PMID: 25481477 PMCID: PMC4358840 DOI: 10.1016/j.ymeth.2014.11.016] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/30/2023] Open
Abstract
During the catabolic process of autophagy, cytoplasmic material is transported to the lysosome for degradation and recycling. This way, autophagy contributes to the homeodynamic turnover of proteins, lipids, nucleic acids, glycogen, and even whole organelles. Autophagic activity is increased by adverse conditions such as nutrient limitation, growth factor withdrawal and oxidative stress, and it generally protects cells and organisms to promote their survival. Misregulation of autophagy is likely involved in numerous human pathologies including aging, cancer, infections and neurodegeneration, so its biomedical relevance explains the still growing interest in this field. Here we discuss the different microscopy-based, biochemical and genetic methods currently available to study autophagy in various tissues of the popular model Drosophila. We show examples for results obtained in different assays, explain how to interpret these with regard to autophagic activity, and how to find out which step of autophagy a given gene product is involved in.
Collapse
|
36
|
Horn L, Leips J, Starz‐Gaiano M. Phagocytic ability declines with age in adult Drosophila hemocytes. Aging Cell 2014; 13:719-28. [PMID: 24828474 PMCID: PMC4116448 DOI: 10.1111/acel.12227] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/17/2022] Open
Abstract
Most multicellular organisms show a physiological decline in immune function with age. However, little is known about the mechanisms underlying these changes. We examined Drosophila melanogaster, an important model for identifying genes affecting innate immunity and senescence, to explore the role of phagocytosis in age-related immune dysfunction. We characterized the localized response of immune cells at the dorsal vessel to bacterial infection in 1-week- and 5-week-old flies. We developed a quantitative phagocytosis assay for adult Drosophila and utilized this to characterize the effect of age on phagocytosis in transgenic and natural variant lines. We showed that genes necessary for bacterial engulfment in other contexts are also required in adult flies. We found that blood cells from young and old flies initially engulf bacteria equally well, while cells from older flies accumulate phagocytic vesicles and thus are less capable of destroying pathogens. Our results have broad implications for understanding how the breakdown in cellular processes influences immune function with age.
Collapse
Affiliation(s)
- Lucas Horn
- Department of Biological Sciences University of Maryland Baltimore County Baltimore MD 21250USA
| | - Jeff Leips
- Department of Biological Sciences University of Maryland Baltimore County Baltimore MD 21250USA
| | - Michelle Starz‐Gaiano
- Department of Biological Sciences University of Maryland Baltimore County Baltimore MD 21250USA
| |
Collapse
|
37
|
Mulakkal NC, Nagy P, Takats S, Tusco R, Juhász G, Nezis IP. Autophagy in Drosophila: from historical studies to current knowledge. BIOMED RESEARCH INTERNATIONAL 2014; 2014:273473. [PMID: 24949430 PMCID: PMC4052151 DOI: 10.1155/2014/273473] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/17/2014] [Indexed: 12/17/2022]
Abstract
The discovery of evolutionarily conserved Atg genes required for autophagy in yeast truly revolutionized this research field and made it possible to carry out functional studies on model organisms. Insects including Drosophila are classical and still popular models to study autophagy, starting from the 1960s. This review aims to summarize past achievements and our current knowledge about the role and regulation of autophagy in Drosophila, with an outlook to yeast and mammals. The basic mechanisms of autophagy in fruit fly cells appear to be quite similar to other eukaryotes, and the role that this lysosomal self-degradation process plays in Drosophila models of various diseases already made it possible to recognize certain aspects of human pathologies. Future studies in this complete animal hold great promise for the better understanding of such processes and may also help finding new research avenues for the treatment of disorders with misregulated autophagy.
Collapse
Affiliation(s)
- Nitha C. Mulakkal
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Peter Nagy
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Szabolcs Takats
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Radu Tusco
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Gábor Juhász
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Ioannis P. Nezis
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
38
|
Feng W, Chang C, Luo D, Su H, Yu S, Hua W, Chen Z, Hu H, Liu W. Dissection of autophagy in human platelets. Autophagy 2014; 10:642-651. [PMID: 24458007 PMCID: PMC4091151 DOI: 10.4161/auto.27832] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 01/18/2023] Open
Abstract
Continuous turnover of intracellular components by autophagy is necessary to preserve cellular homeostasis in all tissues. Despite recent advances in identifying autophagy-related genes and understanding the functions of autophagy in developmental and pathological conditions, so far, the role of autophagy in platelet, a specific anucleate cell type, is poorly understood. In this study, we showed that human platelets express the autophagy-related proteins ATG5, ATG7, and LC3. The same as in nucleated mammalian cells, autophagy was stimulated by cell starvation or the MTOR inhibitor rapamycin in a phosphatidylinositol 3-kinase (PtdIns3K)-dependent manner. Disruption of autophagic flux led to impairment of platelet aggregation and adhesion. Furthermore, Becn1 heterozygous knockout mice displayed a prolonged bleeding time and reduced platelet aggregation. These results suggest a potential role of autophagy in the regulation of platelet function, and imply that gene transcription may not be an essential prerequisite for adaptive autophagy.
Collapse
Affiliation(s)
- Wenfeng Feng
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Chunmei Chang
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Dongjiao Luo
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Hua Su
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Shanshan Yu
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Wen Hua
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| | - Zhihua Chen
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| | - Hu Hu
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Wei Liu
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease; First Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| |
Collapse
|
39
|
Jiang P, Lan Y, Luo J, Ren YL, Liu DG, Pang JX, Liu J, Li J, Wang C, Cai JP. Rapamycin promoted thrombosis and platelet adhesion to endothelial cells by inducing membrane remodeling. BMC Cell Biol 2014; 15:7. [PMID: 24564184 PMCID: PMC3936831 DOI: 10.1186/1471-2121-15-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 02/06/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recently, evidence indicated that the rapamycin-eluting stent which was used worldwide may contribute to an increased risk for thrombosis. On the contrary, other researchers found it was safe. Thus, it is necessary to clarify the effect of rapamycin on thrombosis and the corresponding mechanisms. RESULTS The effects of rapamycin in vivo were evaluated by modified deep vein thrombosis animal model. The platelets were from healthy volunteers and the platelet-endothelium (purchased from ATCC) adhesion in cultured endothelial cells was assessed. Membrane rufflings in endothelial cells were examined by confocal and electron microscope. Thrombus formation increased in rats that were injected with rapamycin. Electron microscope analysis exhibited microvilli on the rapamycin-treated endothelium in rats. Rapamycin enhanced membrane ruffling in human umbilical vein endothelial cells (HUVECs) and adhesion of platelets to HUVECs. The platelet-HUVECs adhesion was attenuated when cells were treated with cytochalacin B. Inhibition of autophagy by 3-methyladenine led to suppression of membrane ruffles in HUVECs and augmentation of platelet-endothelial adhesion. CONCLUSIONS In conclusion, we found that endothelial membrane remodeling induced by rapamycin is crucial for the adhesion of platelets to endothelial cells and thereby for thrombosis in vivo, and that the endothelial membrane remodeling is autophagy dependent.
Collapse
Affiliation(s)
- Ping Jiang
- The Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, No,1, DaHua Road, Dong Dan, Beijing 100730, P,R,China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lammel U, Bechtold M, Risse B, Berh D, Fleige A, Bunse I, Jiang X, Klämbt C, Bogdan S. The Drosophila FHOD1-like formin Knittrig acts through Rok to promote stress fiber formation and directed macrophage migration during the cellular immune response. Development 2014; 141:1366-80. [PMID: 24553290 DOI: 10.1242/dev.101352] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A tight spatiotemporal control of actin polymerization is important for many cellular processes that shape cells into a multicellular organism. The formation of unbranched F-actin is induced by several members of the formin family. Drosophila encodes six formin genes, representing six of the seven known mammalian subclasses. Knittrig, the Drosophila homolog of mammalian FHOD1, is specifically expressed in the developing central nervous system midline glia, the trachea, the wing and in macrophages. knittrig mutants exhibit mild tracheal defects but survive until late pupal stages and mainly die as pharate adult flies. knittrig mutant macrophages are smaller and show reduced cell spreading and cell migration in in vivo wounding experiments. Rescue experiments further demonstrate a cell-autonomous function of Knittrig in regulating actin dynamics and cell migration. Knittrig localizes at the rear of migrating macrophages in vivo, suggesting a cellular requirement of Knittrig in the retraction of the trailing edge. Supporting this notion, we found that Knittrig is a target of the Rho-dependent kinase Rok. Co-expression with Rok or expression of an activated form of Knittrig induces actin stress fibers in macrophages and in epithelial tissues. Thus, we propose a model in which Rok-induced phosphorylation of residues within the basic region mediates the activation of Knittrig in controlling macrophage migration.
Collapse
Affiliation(s)
- Uwe Lammel
- Institute for Neurobiology, University of Münster, 48149 Münster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ishii T, Warabi E, Siow RCM, Mann GE. Sequestosome1/p62: a regulator of redox-sensitive voltage-activated potassium channels, arterial remodeling, inflammation, and neurite outgrowth. Free Radic Biol Med 2013; 65:102-116. [PMID: 23792273 DOI: 10.1016/j.freeradbiomed.2013.06.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
Abstract
Sequestosome1/p62 (SQSTM1) is an oxidative stress-inducible protein regulated by the redox-sensitive transcription factor Nrf2. It is not an antioxidant but known as a multifunctional regulator of cell signaling with an ability to modulate targeted or selective degradation of proteins through autophagy. SQSTM1 implements these functions through physical interactions with different types of proteins including atypical PKCs, nonreceptor-type tyrosine kinase p56(Lck) (Lck), polyubiquitin, and autophagosomal factor LC3. One of the notable physiological functions of SQSTM1 is the regulation of redox-sensitive voltage-gated potassium (Kv) channels which are composed of α and β subunits: (Kvα)4 (Kvβ)4. Previous studies have established that SQSTM1 scaffolds PKCζ, enhancing phosphorylation of Kvβ which induces inhibition of pulmonary arterial Kv1.5 channels under acute hypoxia. Recent studies reveal that Lck indirectly interacts with Kv1.3 α subunits and plays a key role in acute hypoxia-induced Kv1.3 channel inhibition in T lymphocytes. Kv1.3 channels provide a signaling platform to modulate the migration and proliferation of arterial smooth muscle cells and activation of T lymphocytes, and hence have been recognized as a therapeutic target for treatment of restenosis and autoimmune diseases. In this review, we focus on the functional interactions of SQSTM1 with Kv channels through two key partners aPKCs and Lck. Furthermore, we provide molecular insights into the functions of SQSTM1 in suppression of proliferation of arterial smooth muscle cells and neointimal hyperplasia following carotid artery ligation, in T lymphocyte differentiation and activation, and in NGF-induced neurite outgrowth in PC12 cells.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan
| | - Richard C M Siow
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| | - Giovanni E Mann
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| |
Collapse
|
42
|
Chen P, Cescon M, Bonaldo P. Autophagy-mediated regulation of macrophages and its applications for cancer. Autophagy 2013; 10:192-200. [PMID: 24300480 DOI: 10.4161/auto.26927] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved homeostatic pathway that plays an important role in tumor development and progression by acting on cancer cells in a cell-autonomous mechanism. However, the solid tumor is not an island, but rather an ensemble performance that includes nonmalignant stromal cells, such as macrophages. A growing body of evidence indicates that autophagy is a key component of the innate immune response. In this review, we discuss the role of autophagy in the control of macrophage production at different stages (including hematopoietic stem cell maintenance, monocyte/macrophage migration, and monocyte differentiation into macrophages) and polarization and discuss how modulating autophagy in tumor-associated macrophages (TAMs) may represent a promising strategy for limiting cancer growth and progression.
Collapse
Affiliation(s)
- Peiwen Chen
- Department of Molecular Medicine; University of Padova; Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine; University of Padova; Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine; University of Padova; Padova, Italy
| |
Collapse
|
43
|
Belaid A, Ndiaye PD, Cerezo M, Cailleteau L, Brest P, Klionsky DJ, Carle GF, Hofman P, Mograbi B. Autophagy and SQSTM1 on the RHOA(d) again: emerging roles of autophagy in the degradation of signaling proteins. Autophagy 2013; 10:201-8. [PMID: 24300375 DOI: 10.4161/auto.27198] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Degradation of signaling proteins is one of the most powerful tumor-suppressive mechanisms by which a cell can control its own growth, its survival, and its motility. Emerging evidence suggests that autophagy limits several signaling pathways by degrading kinases, downstream components, and transcription factors; however, this often occurs under stressful conditions. Our recent studies revealed that constitutive autophagy temporally and spatially controls the RHOA pathway. Specifically, inhibition of autophagosome degradation induces the accumulation of the GTP-bound form of RHOA. The active RHOA is sequestered via SQSTM1/p62 within autolysosomes, and accordingly fails to localize to the spindle midbody or to the cell surface, as we demonstrate herein. As a result, all RHOA-downstream responses are deregulated, thus driving cytokinesis failure, aneuploidy and motility, three processes that directly have an impact upon cancer progression. We therefore propose that autophagy acts as a degradative brake for RHOA signaling and thereby controls cell proliferation, migration, and genome stability.
Collapse
Affiliation(s)
- Amine Belaid
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | - Papa Diogop Ndiaye
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | - Michaël Cerezo
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; INSERM U895/C3M: Centre Méditerranéen de Médecine Moléculaire; Nice, France
| | - Laurence Cailleteau
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France
| | - Patrick Brest
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | | | - Georges F Carle
- Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Centre Antoine Lacassagne; Nice, France; Laboratoire TIRO-MATOs UMR E4320; Commissariat à l'Energie Atomique; Nice, France
| | - Paul Hofman
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France; Centre Hospitalier Universitaire de Nice; Pasteur Hospital; Laboratory of Clinical and Experimental Pathology; Nice, France
| | - Baharia Mograbi
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| |
Collapse
|
44
|
Bausek N, Zeidler MP. Gα73B is a downstream effector of JAK/STAT signalling and a regulator of Rho1 in Drosophila haematopoiesis. J Cell Sci 2013; 127:101-10. [PMID: 24163435 DOI: 10.1242/jcs.132852] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
JAK/STAT signalling regulates many essential developmental processes including cell proliferation and haematopoiesis, whereas its inappropriate activation is associated with the majority of myeloproliferative neoplasias and numerous cancers. Furthermore, high levels of JAK/STAT pathway signalling have also been associated with enhanced metastatic invasion by cancerous cells. Strikingly, gain-of-function mutations in the single Drosophila JAK homologue, Hopscotch, result in haemocyte neoplasia, inappropriate differentiation and the formation of melanised haemocyte-derived 'tumour' masses; phenotypes that are partly orthologous to human gain-of-function JAK2-associated pathologies. Here we show that Gα73B, a novel JAK/STAT pathway target gene, is necessary for JAK/STAT-mediated tumour formation in flies. In addition, although Gα73B does not affect haemocyte differentiation, it does regulate haemocyte morphology and motility under non-pathological conditions. We show that Gα73B is required for constitutive, but not injury-induced, activation of Rho1 and for the localisation of Rho1 into filopodia upon haemocyte activation. Consistent with these results, we also show that Rho1 interacts genetically with JAK/STAT signalling, and that wild-type levels of Rho1 are necessary for tumour formation. Our findings link JAK/STAT transcriptional outputs, Gα73B activity and Rho1-dependent cytoskeletal rearrangements and cell motility, therefore connecting a pathway associated with cancer with a marker indicative of invasiveness. As such, we suggest a mechanism by which JAK/STAT pathway signalling may promote metastasis.
Collapse
Affiliation(s)
- Nina Bausek
- MRC Centre for Development and Biomedical Genetics, and The Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | | |
Collapse
|
45
|
Scherfer C, Han VC, Wang Y, Anderson AE, Galko MJ. Autophagy drives epidermal deterioration in a Drosophila model of tissue aging. Aging (Albany NY) 2013; 5:276-87. [PMID: 23599123 PMCID: PMC3651520 DOI: 10.18632/aging.100549] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Organismal lifespan has been the primary readout in aging research. However, how longevity genes control tissue-specific aging remains an open question. To examine the crosstalk between longevity programs and specific tissues during aging, biomarkers of organ-specific aging are urgently needed. Since the earliest signs of aging occur in the skin, we sought to examine skin aging in a genetically tractable model. Here we introduce a Drosophila model of skin aging. The epidermis undergoes a dramatic morphological deterioration with age that includes membrane and nuclear loss. These changes were decelerated in a long-lived mutant and accelerated in a short-lived mutant. An increase in autophagy markers correlated with epidermal aging. Finally, the epidermis of Atg7 mutants retained younger characteristics, suggesting that autophagy is a critical driver of epidermal aging. This is surprising given that autophagy is generally viewed as protective during aging. Since Atg7 mutants are short-lived, the deceleration of epidermal aging in this mutant suggests that in the epidermis healthspan can be uncoupled from longevity. Because the aging readout we introduce here has an early onset and is easily visualized, genetic dissection using our model should identify other novel mechanisms by which lifespan genes feed into tissue-specific aging.
Collapse
Affiliation(s)
- Christoph Scherfer
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Unit 1000, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
46
|
Tuloup-Minguez V, Hamaï A, Greffard A, Nicolas V, Codogno P, Botti J. Autophagy modulates cell migration and β1 integrin membrane recycling. Cell Cycle 2013; 12:3317-28. [PMID: 24036548 PMCID: PMC3885642 DOI: 10.4161/cc.26298] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 12/14/2022] Open
Abstract
Cell migration is dependent on a series of integrated cellular events including the membrane recycling of the extracellular matrix receptor integrins. In this paper, we investigate the role of autophagy in regulating cell migration. In a wound-healing assay, we observed that autophagy was reduced in cells at the leading edge than in cells located rearward. These differences in autophagy were correlated with the robustness of MTOR activity. The spatial difference in the accumulation of autophagic structures was not detected in rapamycin-treated cells, which had less migration capacity than untreated cells. In contrast, the knockdown of the autophagic protein ATG7 stimulated cell migration of HeLa cells. Accordingly, atg3(-/-) and atg5(-/-) MEFs have greater cell migration properties than their wild-type counterparts. Stimulation of autophagy increased the co-localization of β1 integrin-containing vesicles with LC3-stained autophagic vacuoles. Moreover, inhibition of autophagy slowed down the lysosomal degradation of internalized β1 integrins and promoted its membrane recycling. From these findings, we conclude that autophagy regulates cell migration, a central mechanism in cell development, angiogenesis, and tumor progression, by mitigating the cell surface expression of β1 integrins.
Collapse
Affiliation(s)
| | - Ahmed Hamaï
- INSERM UMR 845; University of Paris 5 René Descartes; Paris, France
| | - Anne Greffard
- INSERM UMR 984; University of Paris-Sud 11; Châtenay-Malabry, France
| | - Valérie Nicolas
- Microscopy Facility IFR-141-IPSIT; University of Paris-Sud 11; Chatenay-Malabry, France
| | - Patrice Codogno
- INSERM UMR 984; University of Paris-Sud 11; Châtenay-Malabry, France
- INSERM UMR 845; University of Paris 5 René Descartes; Paris, France
| | - Joëlle Botti
- INSERM UMR 984; University of Paris-Sud 11; Châtenay-Malabry, France
- INSERM UMR 845; University of Paris 5 René Descartes; Paris, France
- Lariboisière-Saint Louis Medicine School; University of Paris 7 Denis Diderot; Paris, France
| |
Collapse
|
47
|
Mitochondrial signaling: forwards, backwards, and in between. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:351613. [PMID: 23819011 PMCID: PMC3681274 DOI: 10.1155/2013/351613] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/04/2013] [Indexed: 12/13/2022]
Abstract
Mitochondria are semiautonomous organelles that are a defining characteristic of almost all eukaryotic cells. They are vital for energy production, but increasing evidence shows that they play important roles in a wide range of cellular signaling and homeostasis. Our understanding of nuclear control of mitochondrial function has expanded over the past half century with the discovery of multiple transcription factors and cofactors governing mitochondrial biogenesis. More recently, nuclear changes in response to mitochondrial messaging have led to characterization of retrograde mitochondrial signaling, in which mitochondria have the ability to alter nuclear gene expression. Mitochondria are also integral to other components of stress response or quality control including ROS signaling, unfolded protein response, mitochondrial autophagy, and biogenesis. These avenues of mitochondrial signaling are discussed in this review.
Collapse
|
48
|
Belaid A, Cerezo M, Chargui A, Corcelle-Termeau E, Pedeutour F, Giuliano S, Ilie M, Rubera I, Tauc M, Barale S, Bertolotto C, Brest P, Vouret-Craviari V, Klionsky DJ, Carle GF, Hofman P, Mograbi B. Autophagy plays a critical role in the degradation of active RHOA, the control of cell cytokinesis, and genomic stability. Cancer Res 2013; 73:4311-22. [PMID: 23704209 DOI: 10.1158/0008-5472.can-12-4142] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Degradation of signaling proteins is one of the most powerful tumor-suppressive mechanisms by which a cell can control its own growth. Here, we identify RHOA as the molecular target by which autophagy maintains genomic stability. Specifically, inhibition of autophagosome degradation by the loss of the v-ATPase a3 (TCIRG1) subunit is sufficient to induce aneuploidy. Underlying this phenotype, active RHOA is sequestered via p62 (SQSTM1) within autolysosomes and fails to localize to the plasma membrane or to the spindle midbody. Conversely, inhibition of autophagosome formation by ATG5 shRNA dramatically increases localization of active RHOA at the midbody, followed by diffusion to the flanking zones. As a result, all of the approaches we examined that compromise autophagy (irrespective of the defect: autophagosome formation, sequestration, or degradation) drive cytokinesis failure, multinucleation, and aneuploidy, processes that directly have an impact upon cancer progression. Consistently, we report a positive correlation between autophagy defects and the higher expression of RHOA in human lung carcinoma. We therefore propose that autophagy may act, in part, as a safeguard mechanism that degrades and thereby maintains the appropriate level of active RHOA at the midbody for faithful completion of cytokinesis and genome inheritance.
Collapse
Affiliation(s)
- Amine Belaid
- Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, Laboratoire TIRO-MATOs UMR E4320, Commissariat à l'Energie Atomique, Centre Antoine Lacassagne, Avenue de Valombrose; 06107 Nice Cedex 02, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shravage BV, Hill JH, Powers CM, Wu L, Baehrecke EH. Atg6 is required for multiple vesicle trafficking pathways and hematopoiesis in Drosophila. Development 2013; 140:1321-9. [PMID: 23406899 DOI: 10.1242/dev.089490] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Atg6 (beclin 1 in mammals) is a core component of the Vps34 complex that is required for autophagy. Beclin 1 (Becn1) functions as a tumor suppressor, and Becn1(+/-) tumors in mice possess elevated cell stress and p62 levels, altered NF-κB signaling and genome instability. The tumor suppressor function of Becn1 has been attributed to its role in autophagy, and the potential functions of Atg6/Becn1 in other vesicle trafficking pathways for tumor development have not been considered. Here, we generate Atg6 mutant Drosophila and demonstrate that Atg6 is essential for autophagy, endocytosis and protein secretion. By contrast, the core autophagy gene Atg1 is required for autophagy and protein secretion, but it is not required for endocytosis. Unlike null mutants of other core autophagy genes, all Atg6 mutant animals possess blood cell masses. Atg6 mutants have enlarged lymph glands (the hematopoietic organ in Drosophila), possess elevated blood cell numbers, and the formation of melanotic blood cell masses in these mutants is not suppressed by mutations in either p62 or NFκB genes. Thus, like mammals, altered Atg6 function in flies causes hematopoietic abnormalities and lethality, and our data indicate that this is due to defects in multiple membrane trafficking processes.
Collapse
Affiliation(s)
- Bhupendra V Shravage
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
50
|
Wang BS, Yang Y, Lu HZ, Shang L, Zhang Y, Hao JJ, Shi ZZ, Wang XM, Liu YZ, Zhan QM, Jia XM, Wang MR. Inhibition of atypical protein kinase Cι induces apoptosis through autophagic degradation of β-catenin in esophageal cancer cells. Mol Carcinog 2013; 53:514-25. [PMID: 23359356 DOI: 10.1002/mc.22003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 12/19/2012] [Indexed: 12/21/2022]
Abstract
Atypical protein kinase Cι (PKCι) has been identified as an oncoprotein in esophageal squamous cell carcinomas. However, the mechanisms underlying the role of PKCι in this disease remain unclear. In the present work, we found that inhibition of PKCι expression by RNAi induced apoptosis via the down-regulation of β-catenin in esophageal cancer cells. Furthermore, we found that PKCι regulated β-catenin in an autophagy dependent way. Since down-regulation of β-catenin induced by knockdown of PKCι could be rescued by autophagy inhibition; knockdown of PKCι activated autophagy and promoted the recruitment of β-catenin into autophagosome. These results suggested that PKCι positively regulated β-catenin through negatively regulated autophagy and depletion of PKCι promoted apoptosis via autophagic degradation of β-catenin in esophageal cancer cells. These data provide new insights into PKCι signaling in human cancer.
Collapse
Affiliation(s)
- Bo-Shi Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|