1
|
Tan ZY, Adade JKA, Gu X, Hecht CJS, Salcius M, Tong B, Liu S, Hwang S, Zécri FJ, Graham DB, Schreiber SL, Xavier RJ. Development of an FKBP12-recruiting chemical-induced proximity DNA-encoded library and its application to discover an autophagy potentiator. Cell Chem Biol 2025; 32:498-510.e35. [PMID: 39753134 PMCID: PMC11928285 DOI: 10.1016/j.chembiol.2024.12.002] [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: 05/31/2024] [Revised: 10/01/2024] [Accepted: 12/04/2024] [Indexed: 03/23/2025]
Abstract
Chemical inducers of proximity (CIPs) are molecules that recruit one protein to another and introduce new functionalities toward modulating protein states and activities. While CIP-mediated recruitment of E3 ligases is widely exploited for the development of degraders, other therapeutic modalities remain underexplored. We describe a non-degrader CIP-DNA-encoded library (CIP-DEL) that recruits FKBP12 to target proteins using non-traditional acyclic structures, with an emphasis on introducing stereochemically diverse and rigid connectors to attach the combinatorial library. We deployed this strategy to modulate ATG16L1 T300A, which confers genetic susceptibility to Crohn's disease (CD), and identified a compound that stabilizes the variant protein against caspase-3 (Casp3) cleavage in a FKBP12-independent manner. We demonstrate in cellular models that this compound potentiates autophagy, and reverses the xenophagy defects as well as increased cytokine secretion characteristic of ATG16L1 T300A. This study provides a platform to access unexplored chemical space for CIP design to develop therapeutic modalities guided by human genetics.
Collapse
Affiliation(s)
- Zher Yin Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joel K A Adade
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiebin Gu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cody J S Hecht
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael Salcius
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Bingqi Tong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuang Liu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Seungmin Hwang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Frédéric J Zécri
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Arena BioWorks, Cambridge, MA 02139, USA.
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
2
|
Kaur K, Jewett A. Distinct profiles of osteoclast and dendritic cell-mediated expansion and functional activation of NK and T cells. Cancer Immunol Immunother 2025; 74:127. [PMID: 40024920 PMCID: PMC11872835 DOI: 10.1007/s00262-025-03956-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Osteoclasts (OCs) and dendritic cells (DCs) induce expansion and functional activation of NK and T cells. When comparing OCs with DC-induced activation in NK cells, OCs induced significantly higher cell expansion and functional activation of NK cells as compared to DCs, either from healthy individuals or those obtained from cancer patients. However, no differences could be seen in the levels of cell expansion and functional activation in T cells activated by OCs or DCs, either from healthy individuals or those from cancer patients. OCs selectively expanded and activated CD8 + T cells, whereas DCs expanded and activated CD4 + T cells. In addition, both allogeneic and autologous OCs induced similar levels of cell expansion and functional activation of NK and T cells. Together, these findings highlighted the essential roles of OCs in expanding and activating the cytotoxic effectors of NK, and CD8 + T cells, and demonstrated several differences when compared to the effect of DCs.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
- The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
| |
Collapse
|
3
|
Chakraborty S, Nandi P, Mishra J, Niharika, Roy A, Manna S, Baral T, Mishra P, Mishra PK, Patra SK. Molecular mechanisms in regulation of autophagy and apoptosis in view of epigenetic regulation of genes and involvement of liquid-liquid phase separation. Cancer Lett 2024; 587:216779. [PMID: 38458592 DOI: 10.1016/j.canlet.2024.216779] [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: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
Cellular physiology is critically regulated by multiple signaling nexuses, among which cell death mechanisms play crucial roles in controlling the homeostatic landscape at the tissue level within an organism. Apoptosis, also known as programmed cell death, can be induced by external and internal stimuli directing the cells to commit suicide in unfavourable conditions. In contrast, stress conditions like nutrient deprivation, infection and hypoxia trigger autophagy, which is lysosome-mediated processing of damaged cellular organelle for recycling of the degraded products, including amino acids. Apparently, apoptosis and autophagy both are catabolic and tumor-suppressive pathways; apoptosis is essential during development and cancer cell death, while autophagy promotes cell survival under stress. Moreover, autophagy plays dual role during cancer development and progression by facilitating the survival of cancer cells under stressed conditions and inducing death in extreme adversity. Despite having two different molecular mechanisms, both apoptosis and autophagy are interconnected by several crosslinking intermediates. Epigenetic modifications, such as DNA methylation, post-translational modification of histone tails, and miRNA play a pivotal role in regulating genes involved in both autophagy and apoptosis. Both autophagic and apoptotic genes can undergo various epigenetic modifications and promote or inhibit these processes under normal and cancerous conditions. Epigenetic modifiers are uniquely important in controlling the signaling pathways regulating autophagy and apoptosis. Therefore, these epigenetic modifiers of both autophagic and apoptotic genes can act as novel therapeutic targets against cancers. Additionally, liquid-liquid phase separation (LLPS) also modulates the aggregation of misfolded proteins and provokes autophagy in the cytosolic environment. This review deals with the molecular mechanisms of both autophagy and apoptosis including crosstalk between them; emphasizing epigenetic regulation, involvement of LLPS therein, and possible therapeutic approaches against cancers.
Collapse
Affiliation(s)
- Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Prahallad Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bypass Road, Bhauri, Bhopal, 462 030, MP, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India.
| |
Collapse
|
4
|
Kaur K, Jewett A. Similarities and Differences between Osteoclast-Mediated Functional Activation of NK, CD3+ T, and γδ T Cells from Humans, Humanized-BLT Mice, and WT Mice. Crit Rev Immunol 2024; 44:61-75. [PMID: 38305337 DOI: 10.1615/critrevimmunol.2023051091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
This study is focused on assessing the activation in NK, CD3+ T, and γδ T cells when they interact with osteoclasts (OCs) and monocytes in the presence or absence of zoledronate (ZOL), both in humans and WT mice. OCs resulted in increased IFN-γ secretion in NK, CD3+ T, and γδ T cells, however, the significantly highest increase was seen when cells were co-cultured with ZOL-treated OCs. Our previous studies have demonstrated increased IFN-γ secretion in the peripheral blood-derived immune cells of bisphosphonate-related osteonecrosis of the jaw (BRONJ) mice model. This could be due to increased OCs-induced activation of immune cells with ZOL treatment. We also observed increased IFN-γ secretion in humanized-BLT (hu-BLT) mice NK cells when were co-cultured with OCs or monocytes, and higher IFN-γ secretion levels were seen in the presence of OCs or ZOL-treated OCs. In addition, similar effects on IFN-γ secretion levels of NK, CD3+ T, and γδ T cells were seen whether cells were co-cultured with allogeneic OCs or autologous OCs.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA; The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA
| |
Collapse
|
5
|
Chen PC, Kaur K, Ko MW, Huerta-Yepez S, Jain Y, Jewett A. Regulation of Cytotoxic Immune Effector Function by AJ3 Probiotic Bacteria in Amyotrophic Lateral Sclerosis (ALS). Crit Rev Immunol 2023; 43:13-26. [PMID: 37522558 DOI: 10.1615/critrevimmunol.2023047231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Our recent studies indicated that amyotrophic lateral sclerosis (ALS) patients suffer from significantly elevated levels of interferon-gamma (IFN-γ) secretion by natural killer (NK) and CD8+ T cells, which may be responsible for the immune-pathologies seen in central nervous system and in peripheral organs of the patients. In order to counter such elevated induction of IFN-γ in patients we designed a treatment strategy to increase anti-inflammatory cytokine interleukin-10 (IL-10) by the use of probiotic strains which significantly increase the levels of IL-10. Therefore, in this paper we demonstrate disease specific functions of Al-Pro (AJ3) formulated for the adjunct treatment of auto-immune diseases including ALS, and compared the function with CA/I-Pro (AJ4) for the treatment of cancer and viral diseases, and NK-CLK (AJ2) for maintenance of immune balance and promotion of disease prevention. The three different formulations of probiotic bacteria have distinct profiles of activation of peripheral blood mononuclear cells (PBMCs), NK, and CD8+ T cells, and their induced activation is different from those mediated by either IL-2 or IL-2 + anti-CD16 monoclonal antibodies (mAbs) or IL-2 + anti-CD3/CD28 mAbs. IL-2 + anti-CD16 mAb activation of PBMCs and NK cells had the highest IFN-γ/IL-10 ratio, whereas IL-2 combination with sAJ4 had the next highest followed by IL-2 + sAJ2 and the lowest was seen with IL-2 + sAJ3. Accordingly, the highest secretion of IFN-γ was seen when the PBMCs and NK cells were treated with IL-2 + sAJ4, intermediate for IL-2 + sAJ2 and the lowest with IL-2 + sAJ3. The levels of IFN-γ induction and the ratio of IFN-γ to IL-10 induced by different probiotic bacteria formulation in the absence of IL-2 treatment remained much lower when compared to those treated in the presence of IL-2. Of note is the difference between NK cells and CD8+ T cells in which synergistic induction of IFN-y by IL-2 + sAJ4 was significantly higher in NK cells than those seen by CD8+ T cells. Based on these results, sAJ3 should be effective in alleviating auto-immunity seen in ALS since it will greatly regulate the levels and function of IFN-γ negatively, decreasing overactivation of cytotoxic immune effectors and prevention of death in motor neurons.
Collapse
Affiliation(s)
- Po-Chun Chen
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Meng-Wei Ko
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Sara Huerta-Yepez
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, Los Angeles, CA 90095, USA
| | - Yash Jain
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, Los Angeles, CA 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA; The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA
| |
Collapse
|
6
|
Triantos C, Aggeletopoulou I, Mantzaris GJ, Mouzaki Α. Molecular basis of vitamin D action in inflammatory bowel disease. Autoimmun Rev 2022; 21:103136. [DOI: 10.1016/j.autrev.2022.103136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
|
7
|
Zhang C, Wang X, Sun J, Guo M, Zhang X, Wu Y. Autophagy Induced by the N-Terminus of the Classic Swine Fever Virus Nonstructural Protein 5A Protein Promotes Viral Replication. Front Microbiol 2021; 12:733385. [PMID: 34512612 PMCID: PMC8424089 DOI: 10.3389/fmicb.2021.733385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/31/2021] [Indexed: 12/13/2022] Open
Abstract
Although classic swine fever virus (CSFV) infection has been reported to induce autophagy, the specific induced mechanism remains unrevealed. Nonstructural protein 5A (NS5A) of CSFV is a multiphosphorylated protein with multiple functions to regulate viral replication and the host cell immune responses. Herein, we demonstrated that CSFV NS5A could induce cellular autophagy and promote viral replication. In the current study, we showed that NS5A expression significantly increased the levels of autophagy-related genes (ATGs), including light chain 3 (LC3), ATG5, and Beclin 1; conversely, degradation of P62/sequestosome 1 (SQSTM1) was observed by Western blotting. The number of autophagy-like vesicles was also obviously increased in NS5A-expressing cells, as analyzed by transmission electron microscopy (TEM). Furthermore, we observed the co-localization of the NS5A and LC3 proteins by confocal immunofluorescence analysis. Direct binding of NS5A to the autophagy-related LC3 protein was confirmed by coimmunoprecipitation in vivo and by a GST pulldown assay in vitro. Through segmentation and point mutation research on the NS5A protein, we found that the N-terminal region and the phosphorylation of amino acids 81 and 92 of the NS5A protein were essential for inducing autophagy. Finally, we demonstrated that the LC3 protein had a positive effect on CSFV replication. These findings emphasize a previously unascertained interaction relationship between NS5A and LC3 in the autophagy process. Furthermore, our research revealed a new role of CSFV NS5A, particularly its N-terminal amino acids serine 81 and serine 92, as a critical regulator of CSFV-induced autophagy and have significance for extending our understanding of the CSFV-autophagy interplay.
Collapse
Affiliation(s)
- Chengcheng Zhang
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiuling Wang
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jiahao Sun
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| | - Mengjiao Guo
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaorong Zhang
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yantao Wu
- College of Veterinary Medicine, Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
8
|
Mitochondrial Modulations, Autophagy Pathways Shifts in Viral Infections: Consequences of COVID-19. Int J Mol Sci 2021; 22:ijms22158180. [PMID: 34360945 PMCID: PMC8347486 DOI: 10.3390/ijms22158180] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/24/2021] [Accepted: 07/24/2021] [Indexed: 01/05/2023] Open
Abstract
Mitochondria are vital intracellular organelles that play an important role in regulating various intracellular events such as metabolism, bioenergetics, cell death (apoptosis), and innate immune signaling. Mitochondrial fission, fusion, and membrane potential play a central role in maintaining mitochondrial dynamics and the overall shape of mitochondria. Viruses change the dynamics of the mitochondria by altering the mitochondrial processes/functions, such as autophagy, mitophagy, and enzymes involved in metabolism. In addition, viruses decrease the supply of energy to the mitochondria in the form of ATP, causing viruses to create cellular stress by generating ROS in mitochondria to instigate viral proliferation, a process which causes both intra- and extra-mitochondrial damage. SARS-COV2 propagates through altering or changing various pathways, such as autophagy, UPR stress, MPTP and NLRP3 inflammasome. Thus, these pathways act as potential targets for viruses to facilitate their proliferation. Autophagy plays an essential role in SARS-COV2-mediated COVID-19 and modulates autophagy by using various drugs that act on potential targets of the virus to inhibit and treat viral infection. Modulated autophagy inhibits coronavirus replication; thus, it becomes a promising target for anti-coronaviral therapy. This review gives immense knowledge about the infections, mitochondrial modulations, and therapeutic targets of viruses.
Collapse
|
9
|
Eva1a inhibits NLRP3 activation to reduce liver ischemia-reperfusion injury via inducing autophagy in kupffer cells. Mol Immunol 2021; 132:82-92. [PMID: 33556710 DOI: 10.1016/j.molimm.2021.01.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/03/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
Ischemia-reperfusion(IR) injury is one of the main complications of liver transplantation and partial hepatectomy. Innate immunity mediated by kupffer cells plays an important role in it. In this study, we focused on evaluating the intrinsic relationship between the autophagy induction of kupffer cells and the activation of NLRP3 inflammasomes caused by liver ischemia-reperfusion. Pre-depletion of kupffer cells can aggravate inflammation and tissue damage within 24 h after IR.Enhancing the autophagy of kupffer cells can inhibit the activation of NLRP3 caused by IR, and inhibiting autophagy can induce the secretion of IL1β dependent on NLRP3 activation.Eva1a is up-regulated by the inflammatory cascade activated by IR.Knockdown of Eva1a in vivo on the one hand will aggravate IR inflammation, increase the production of TNF-α, IL-1β and inhibit the secretion of IL-10.On the other hand, it will aggravate the liver histological damage. Knockout of Eva1a induces ASC activation and cleavage of caspase1 and IL1β in an NLRP3-dependent manner, which is closely related to the function of blocking Eva1a to promote autophagosome formation.We further found that knockdown of ATG16L1 will reverse the more formation of autophagosomes induced by overexpression of Eva1a, whereas knockdown of ATG16L1 did not further reduce the formation of autophagosomes inhibited by siEva1a. We also found that the addition of siATG7, siATG5 and siATG12 would reverse the IR autophagy of liver induced by overexpression of Eva1a, but inhibition of the Beclin1-Vps34 pathway did not significantly reverse the effect of overexpression of Eva1a.These prove that Eva1a and ATG16L1 may work together in the liver IR model to actively induce the formation of autophagosomes and be independent from the beclin1-vps34-induced autophagy pathway to limit the excessive activation of IR inflammation. Our study provides brand new insights into the mechanism of liver macrophages in the progression of inflammation in the context of liver ischemia-reperfusion injury.
Collapse
|
10
|
Crezee T, Tesselaar MH, Nagarajah J, Corver WE, Morreau J, Pritchard C, Kimura S, Kuiper JG, van Engen-van Grunsven I, Smit JWA, Netea-Maier RT, Plantinga TS. Digoxin treatment reactivates in vivo radioactive iodide uptake and correlates with favorable clinical outcome in non-medullary thyroid cancer. Cell Oncol (Dordr) 2021; 44:611-625. [PMID: 33534128 PMCID: PMC8213564 DOI: 10.1007/s13402-021-00588-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Non-medullary thyroid cancer (NMTC) treatment is based on the ability of thyroid follicular cells to accumulate radioactive iodide (RAI). However, in a subset of NMTC patients tumor dedifferentiation occurs, leading to RAI resistance. Digoxin has been demonstrated to restore iodide uptake capacity in vitro in poorly differentiated and anaplastic NMTC cells, termed redifferentiation. The aim of the present study was to investigate the in vivo effects of digoxin in TPO-Cre/LSL-BrafV600E mice and digoxin-treated NMTC patients. METHODS Mice with thyroid cancer were subjected to 3D ultrasound for monitoring tumor growth and 124I PET/CT for measurement of intratumoral iodide uptake. Post-mortem analyses on tumor tissues comprised gene expression profiling and measurement of intratumoral autophagy activity. Through PALGA (Dutch Pathology Registry), archived tumor material was obtained from 11 non-anaplastic NMTC patients who were using digoxin. Clinical characteristics and tumor material of these patients were compared to 11 matched control NMTC patients never treated with digoxin. RESULTS We found that in mice, tumor growth was inhibited and 124I accumulation was sustainably increased after short-course digoxin treatment. Post-mortem analyses revealed that digoxin treatment increased autophagy activity and enhanced expression of thyroid-specific genes in mouse tumors compared to vehicle-treated mice. Digoxin-treated NMTC patients exhibited significantly higher autophagy activity and a higher differentiation status as compared to matched control NMTC patients, and were associated with favourable clinical outcome. CONCLUSIONS These in vivo data support the hypothesis that digoxin may represent a repositioned adjunctive treatment modality that suppresses tumor growth and improves RAI sensitivity in patients with RAI-refractory NMTC.
Collapse
Affiliation(s)
- Thomas Crezee
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands.
| | - Marika H Tesselaar
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| | - James Nagarajah
- Department of Radiology & Nuclear Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Willem E Corver
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catrin Pritchard
- Department of Pathology, University of Leicester, Leicester, LEI7RH, UK
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Ilse van Engen-van Grunsven
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| | - Jan W A Smit
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Theo S Plantinga
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
11
|
Mizushima N, Murphy LO. Autophagy Assays for Biological Discovery and Therapeutic Development. Trends Biochem Sci 2020; 45:1080-1093. [DOI: 10.1016/j.tibs.2020.07.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/08/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022]
|
12
|
Kaur K, Ko MW, Ohanian N, Cook J, Jewett A. Osteoclast-expanded super-charged NK-cells preferentially select and expand CD8+ T cells. Sci Rep 2020; 10:20363. [PMID: 33230147 PMCID: PMC7683603 DOI: 10.1038/s41598-020-76702-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoclasts (OCs) and much less dendritic cells (DCs) induce significant expansion and functional activation of NK cells, and furthermore, the OC-expanded NK cells preferentially increase the expansion and activation of CD8+ T cells by targeting CD4+ T cells. When autologous OCs were used to expand patient NK cells much lower percentages of expanded CD8+ T cells, decreased numbers of expanded NK cells and decreased functions of NK cells could be observed, and the addition of allogeneic healthy OCs increased the patients' NK function. Mechanistically, OC-expanded NK cells were found to lyse CD4+ T cells but not CD8+ T cells suggesting potential selection of CD8+ T cells before their expansion by OC activated NK cells. In agreement, Increased IFN-γ secretion, and NK cell-mediated cytotoxicity and higher percentages of CD8+ T cells, in various tissue compartments of oral tumor-bearing hu-BLT mice in response to immunotherapy by OC-expanded NK cells were observed. Thus, our results indicate an important relationship between NK and CD8+ T cells.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Meng-Wei Ko
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Nick Ohanian
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Jessica Cook
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA.
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.
- The Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Hassanpour M, Hajihassani F, Hiradfar A, Aghamohammadzadeh N, Rahbarghazi R, Safaie N, Nouri M, Panahi Y. Real-state of autophagy signaling pathway in neurodegenerative disease; focus on multiple sclerosis. JOURNAL OF INFLAMMATION-LONDON 2020; 17:6. [PMID: 32082082 PMCID: PMC7014934 DOI: 10.1186/s12950-020-0237-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
Abstract
The occurrence of neurodegenerative disease is increasingly raised. From physiopathological aspect, the emergence of auto-reactive antibodies against the nervous system antigens contributes to de-myelination in Multiple sclerosis (MS). These features cause the nervous system dysfunction. The follow-up of molecular alterations could give us a real-state vision about intracellular status during pathological circumstances. In this review, we focus on the autophagic response during MS progression and further understand the relationship between autophagy and MS and its modulatory effect on the MS evolution. The authors reviewed studies published on the autophagy status in neurodegenerative disease and on the autophagy modulation in MS prognosis, diagnosis, and possible therapies. The inevitable role of autophagy was shown in the early-stage progression of MS. Due to critical role of autophagy in different stage of cell activity in nervous system, the distinct role of autophagy should not be neglected in the development, pathogenesis, and treatment of MS.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- 1Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, 5166614756, Imam Reza St., Golgasht St, Tabriz, Iran.,2Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,3Clinical Pharmacy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, 1435916471 Iran
| | - Fateme Hajihassani
- 4Department of Health Management, School of Management and Medical informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirataollah Hiradfar
- 5Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- 7Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,8Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Safaie
- 9Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- 1Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, 5166614756, Imam Reza St., Golgasht St, Tabriz, Iran.,2Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yunes Panahi
- 3Clinical Pharmacy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, 1435916471 Iran
| |
Collapse
|
14
|
Goes A, Lapuhs P, Kuhn T, Schulz E, Richter R, Panter F, Dahlem C, Koch M, Garcia R, Kiemer AK, Müller R, Fuhrmann G. Myxobacteria-Derived Outer Membrane Vesicles: Potential Applicability Against Intracellular Infections. Cells 2020; 9:cells9010194. [PMID: 31940898 PMCID: PMC7017139 DOI: 10.3390/cells9010194] [Citation(s) in RCA: 28] [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: 12/06/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
In 2019, it was estimated that 2.5 million people die from lower tract respiratory infections annually. One of the main causes of these infections is Staphylococcus aureus, a bacterium that can invade and survive within mammalian cells. S. aureus intracellular infections are difficult to treat because several classes of antibiotics are unable to permeate through the cell wall and reach the pathogen. This condition increases the need for new therapeutic avenues, able to deliver antibiotics efficiently. In this work, we obtained outer membrane vesicles (OMVs) derived from the myxobacteria Cystobacter velatus strain Cbv34 and Cystobacter ferrugineus strain Cbfe23, that are naturally antimicrobial, to target intracellular infections, and investigated how they can affect the viability of epithelial and macrophage cell lines. We evaluated by cytometric bead array whether they induce the expression of proinflammatory cytokines in blood immune cells. Using confocal laser scanning microscopy and flow cytometry, we also investigated their interaction and uptake into mammalian cells. Finally, we studied the effect of OMVs on planktonic and intracellular S. aureus. We found that while Cbv34 OMVs were not cytotoxic to cells at any concentration tested, Cbfe23 OMVs affected the viability of macrophages, leading to a 50% decrease at a concentration of 125,000 OMVs/cell. We observed only little to moderate stimulation of release of TNF-alpha, IL-8, IL-6 and IL-1beta by both OMVs. Cbfe23 OMVs have better interaction with the cells than Cbv34 OMVs, being taken up faster by them, but both seem to remain mostly on the cell surface after 24 h of incubation. This, however, did not impair their bacteriostatic activity against intracellular S. aureus. In this study, we provide an important basis for implementing OMVs in the treatment of intracellular infections.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Philipp Lapuhs
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Thomas Kuhn
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Eilien Schulz
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Robert Richter
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Drug Delivery (DDEL), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Fabian Panter
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Marcus Koch
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany;
| | - Ronald Garcia
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Rolf Müller
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
| | - Gregor Fuhrmann
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Correspondence: ; Tel.: +49-68-198-806 (ext. 1500)
| |
Collapse
|
15
|
Mercado N, Colley T, Baker JR, Vuppussetty C, Kono Y, Clarke C, Tooze S, Johansen T, Barnes PJ. Bicaudal D1 impairs autophagosome maturation in chronic obstructive pulmonary disease. FASEB Bioadv 2019; 1:688-705. [PMID: 32123815 PMCID: PMC6996363 DOI: 10.1096/fba.2018-00055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 11/20/2018] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Bicaudal D1 (BICD1), an adaptor for the dynein-dynactin motor complex, has been identified as a susceptibility gene in chronic obstructive pulmonary disease (COPD). Autophagy, an essential cellular homeostasis process, is defective in COPD, in which oxidative stress-induced misfolded proteins accumulate into toxic aggregates dependent on the accumulation of the autophagic cargo receptor p62. Defective autophagy can be caused by mutations in the dynein and dynactin motor complex suggesting a possible link between BICD1 and defective autophagy in COPD. BICD1 levels were measured in peripheral lung tissue from COPD patients together with markers of autophagy and found to be increased in COPD together with autophagosomes, p62 and p62 oligomers. In vitro exposure of bronchial epithelial cells to cigarette smoke extracts (CSEs) revealed that high concentrations of CSE induced defective autophagosome maturation with accumulation of BICD1, p62 and ubiquitin-associated p62 oligomers. This was confirmed in vivo using CS-exposed mice. Furthermore, we identified that formation of CS-induced p62 oligomers required an interaction with Keap1. Overexpression and ablation of BICD1 confirmed that increased BICD1 negatively regulates autophagosome maturation inducing accumulation of p62 and p62 oligomers and that it can be reversed by cardiac glycosides. We conclude that defective autophagosome maturation in COPD is caused by oxidative stress-mediated BICD1 accumulation.
Collapse
Affiliation(s)
- Nicolas Mercado
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
- Novartis Institutes for BioMedical ResearchBaselSwitzerland
| | - Thomas Colley
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
- Pulmocide LtdLondonUK
| | - Jonathan R. Baker
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | | | - Yuta Kono
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Colin Clarke
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Sharon Tooze
- London Research InstituteCancer Research UKLondonUK
| | - Terje Johansen
- Molecular Cancer Research GroupInstitute of Medical BiologyUniversity of Tromsø – The Arctic University of NorwayTromsøNorway
| | - Peter J. Barnes
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| |
Collapse
|
16
|
Abstract
Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.
Collapse
Affiliation(s)
- Holm H Uhlig
- Department of Pediatrics, University of Oxford, Oxford OX3 9DU, United Kingdom; .,Translational Gastroenterology Unit, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom; .,Translational Gastroenterology Unit, University of Oxford, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
17
|
Zhang Q, Fang X, Zhao W, Liang Q. The transcriptional coactivator YAP1 is overexpressed in osteoarthritis and promotes its progression by interacting with Beclin-1. Gene 2019; 689:210-219. [PMID: 30496783 DOI: 10.1016/j.gene.2018.11.068] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/17/2018] [Accepted: 11/20/2018] [Indexed: 01/06/2023]
Abstract
Osteoarthritis (OA) constitutes the most common disease of degenerative joints, with chondrocytes playing an important role in disease progression. However, the underlying pathobiological mechanisms have not been fully characterized. In this study, we investigated the role of Yes-associated protein 1 (YAP1)-regulated autophagy in chondrocyte proliferation, apoptosis, and differentiation. The data showed that YAP1, a transcriptional coactivator, was overexpressed in OA tissues from a murine model of OA, as analyzed by real time PCR and western blot. Overexpression of YAP1 significantly suppressed ATDC5 chondrogenic cell proliferation and decreased the expression of differentiation-related genes including Runx2, osteocalcin, and collagen I, and elevated cell apoptosis, whereas these cellular processes were reversed by knockdown of YAP1. Immunofluorescence analysis demonstrated that YAP1 co-localized with the autophagy regulator beclin1. Co-immunoprecipitation experiments indicated that this interaction was enhanced in OA tissues. In contrast, YAP1 lacking the internal WW domains failed to interact with beclin1 and was unable to inhibit beclin1 ubiquitination. This resulted in upregulated autophagy, which significantly improved OA by increasing chondrocyte proliferation and differentiation. Notably, YAP1 expression was significantly downregulated by various anti-OA drugs. Finally, the Yap1 promoter was activated by transcriptional factors AP2α and SP1, whereas its 3'UTR was targeted by miR-5624-5p, miR-33-3p, and miR-6918-5p. In conclusion, inhibition of YAP1 could facilitate beclin1-regulated autophagy in OA, suggesting a potential therapeutic approach to combat OA.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning 110000, PR China; Department of Orthopedics, Benxi Central Hospital of China Medical University, Benxi, Liaoning, PR China
| | - Xin Fang
- Department of Orthopedics, Benxi Central Hospital of China Medical University, Benxi, Liaoning, PR China
| | - Wei Zhao
- Department of Clinical Biochemistry, School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Qingwei Liang
- Department of Sports Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
18
|
Autophagy in regulatory T cells: A double-edged sword in disease settings. Mol Immunol 2019; 109:43-50. [PMID: 30852245 DOI: 10.1016/j.molimm.2019.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/03/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is an evolutionarily conserved catabolic process that directs cytoplasmic proteins, organelles and microbes to lysosomes for degradation. It not only represents an essential cell-intrinsic mechanism to protect against internal and external stresses but also shapes both innate and adaptive immunity. Regulatory T cells (Tregs) are a developmentally and functionally distinct T cell subpopulation engaged in sustaining immunological self-tolerance and homeostasis. There is compelling evidence that autophagy is actively regulated in Tregs and serves as a central signal-dependent controller for Tregs by restraining excessive apoptotic and metabolic activities. In this review, we discuss how autophagy modulates the stability and functionality of Tregs in different disease settings, and provide a perspective on how manipulation of autophagy enables better control of immune response by targeting the generation of Tregs and the maintenance of their stability.
Collapse
|
19
|
Renga G, Oikonomou V, Stincardini C, Pariano M, Borghi M, Costantini C, Bartoli A, Garaci E, Goldstein AL, Romani L. Thymosin β4 limits inflammation through autophagy. Expert Opin Biol Ther 2019; 18:171-175. [PMID: 30063848 DOI: 10.1080/14712598.2018.1473854] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Thymosin β4 (Tβ4) is a thymic hormone with multiple and different intracellular and extracellular activities affecting wound healing, inflammation, fibrosis and tissue regeneration. As the failure to resolve inflammation leads to uncontrolled inflammatory pathology which underlies many chronic diseases, the endogenous pathway through which Tβ4 may promote inflammation resolution becomes of great interest. In this review, we discuss data highlighting the efficacy of Tβ4 in resolving inflammation by restoring autophagy. AREAS COVERED The authors provide an overview of the Tβ4's anti-inflammatory properties in several pathologies and provide preliminary evidence on the ability of Tβ4 to resolve inflammation via the promotion of non-canonical autophagy associated with the activation of the DAP kinase anti-inflammatory function. EXPERT OPINION Based on its multitasking activity in various animal studies, including tissue repair and prevention of chronic inflammation, Tβ4 may represent a potential, novel treatment for inflammatory diseases associated with defective autophagy.
Collapse
Affiliation(s)
- Giorgia Renga
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Vasilis Oikonomou
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Claudia Stincardini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Marilena Pariano
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Monica Borghi
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Claudio Costantini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Andrea Bartoli
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Enrico Garaci
- b University San Raffaele and IRCCS San Raffaele , Rome , Italy
| | - Allan L Goldstein
- c Department of Biochemistry and Molecular Medicine , the George Washington University, School of Medicine and Health Sciences , Washington , DC , USA
| | - Luigina Romani
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| |
Collapse
|
20
|
Liguori M, Nuzziello N, Licciulli F, Consiglio A, Simone M, Viterbo RG, Creanza TM, Ancona N, Tortorella C, Margari L, Grillo G, Giordano P, Liuni S, Trojano M. Combined microRNA and mRNA expression analysis in pediatric multiple sclerosis: an integrated approach to uncover novel pathogenic mechanisms of the disease. Hum Mol Genet 2019; 27:66-79. [PMID: 29087462 DOI: 10.1093/hmg/ddx385] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a complex disease of the CNS that usually affects young adults, although 3-5% of cases are diagnosed in childhood and adolescence (hence called pediatric MS, PedMS). Genetic predisposition, among other factors, seems to contribute to the risk of the onset, in pediatric as in adult ages, but few studies have investigated the genetic 'environmentally naïve' load of PedMS. The main goal of this study was to identify circulating markers (miRNAs), target genes (mRNAs) and functional pathways associated with PedMS; we also verified the impact of miRNAs on clinical features, i.e. disability and cognitive performances. The investigation was performed in 19 PedMS and 20 pediatric controls (PCs) using a High-Throughput Next-generation Sequencing (HT-NGS) approach followed by an integrated bioinformatics/biostatistics analysis. Twelve miRNAs were significantly upregulated (let-7a-5p, let-7b-5p, miR-25-3p, miR-125a-5p, miR-942-5p, miR-221-3p, miR-652-3p, miR-182-5p, miR-185-5p, miR-181a-5p, miR-320a, miR-99b-5p) and 1 miRNA was downregulated (miR-148b-3p) in PedMS compared with PCs. The interactions between the significant miRNAs and their targets uncovered predicted genes (i.e. TNFSF13B, TLR2, BACH2, KLF4) related to immunological functions, as well as genes involved in autophagy-related processes (i.e. ATG16L1, SORT1, LAMP2) and ATPase activity (i.e. ABCA1, GPX3). No significant molecular profiles were associated with any PedMS demographic/clinical features. Both miRNAs and mRNA expressions predicted the phenotypes (PedMS-PC) with an accuracy of 92% and 91%, respectively. In our view, this original strategy of contemporary miRNA/mRNA analysis may help to shed light in the genetic background of the disease, suggesting further molecular investigations in novel pathogenic mechanisms.
Collapse
Affiliation(s)
- Maria Liguori
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Nicoletta Nuzziello
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy.,Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Flavio Licciulli
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Arianna Consiglio
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Marta Simone
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Rosa Gemma Viterbo
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Teresa Maria Creanza
- National Research Council of Italy, Department of Engineering, ICT and Technology for Energy and Transportation, Institute of Intelligent Systems for Automation, 70125 Bari, Italy
| | - Nicola Ancona
- National Research Council of Italy, Department of Engineering, ICT and Technology for Energy and Transportation, Institute of Intelligent Systems for Automation, 70125 Bari, Italy
| | - Carla Tortorella
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy.,Department of Neurosciences, San Camillo Forlanini Hospital, 00185 Rome, Italy
| | - Lucia Margari
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Giorgio Grillo
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Paola Giordano
- General Paediatric Unit "B. Trambusti", Azienda Policlinico-Giovanni XXIII and University of Bari, 70125 Bari, Italy
| | - Sabino Liuni
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Maria Trojano
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| |
Collapse
|
21
|
Xiao X, Xu M, Yang C, Yao Y, Liang LN, Ed Chung P, Long Q, Zacksenhaus E, He Z, Liu S, Ben-David Y. Novel racemosin B derivatives as new therapeutic agents for aggressive breast cancer. Bioorg Med Chem 2018; 26:6096-6104. [PMID: 30471828 DOI: 10.1016/j.bmc.2018.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 02/01/2023]
Abstract
Carbazole derivatives show anti-cancer activity and are of great interest for drug development. In this study, we synthesized and analyzed several new alkylamide derivatives of racemocin B, a natural indolo[3,2-a]carbazole molecule originally isolated from the green alga Caulerpa racemose. Several alkylamide derivatives were found to exhibit moderate to strong growth inhibition against human breast cancer cell lines. They induced G2/M cell cycle arrest and apoptosis in the aggressive triple-negative breast cancer cell line MDA-MB-231. Among these derivatives, compound 25 with the lowest IC50 induced cell death by suppressing autophagy. This was accompanied by inhibition of autophagic flux and accumulation of autophagy protein 1 light chain 3, LC3II, and p62. The novel alkylamide derivative offers a potential new treatment for human breast cancer.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China
| | - Mei Xu
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Chao Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Yao Yao
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Li-Na Liang
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Philip Ed Chung
- Department of Medicine, University of Toronto, Toronto, Ontario M5G2M1, Canada; Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, Ontario M5G2M1, Canada
| | - Qun Long
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China
| | - Eldad Zacksenhaus
- Department of Medicine, University of Toronto, Toronto, Ontario M5G2M1, Canada; Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, Ontario M5G2M1, Canada
| | - Zhixu He
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China.
| | - Sheng Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| |
Collapse
|
22
|
Orsatti CL, Sobreira ML, Sandrim VC, Nahas-Neto J, Orsatti FL, Nahas EAP. Autophagy-related 16-like 1gene polymorphism, risk factors for cardiovascular disease and associated carotid intima-media thickness in postmenopausal women. Clin Biochem 2018; 61:12-17. [PMID: 30236831 DOI: 10.1016/j.clinbiochem.2018.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Early identification of asymptomatic postmenopausal women (PW), who are more predisposed to developing cardiovascular disease (CVD), is an important preventive strategy. Autophagy-related 16-like 1 (ATG16L1) is an autophagy gene known to control host immune responses and is associated with a variety of diseases, including CVD. OBJECTIVE The aim of the study was to associate the ATG16L1 polymorphism variant with subclinical carotid atherosclerosis in asymptomatic PW. STUDY DESIGN This cross-sectional study included 210 Brazilian postmenopausal women (age ≥ 45 years with amenorrhea ≥12 months). Clinical, anthropometric and biochemical assessments were performed to evaluate the cardiovascular risk factors. DNA was extracted from buccal cells and the ATG16L1 (T300A) polymorphism was determined by the polymerase chain reaction (PCR). The carotid intima-media thickness and/or the presence of plaques were evaluated by carotid duplex ultrasound. For statistical analysis, the t-test, logistic regression and analysis of covariance (ANCOVA) were used. RESULTS The presence of the polymorphic allele forATG16L1 (T300A) was found in 77.47% (A/G = 49.87%, G/G = 27.60%). The ATG16L1 (T300A) polymorphism is significantly associated with increased carotid intima-media thickness (IMT) after adjustments of the confounding variables (P < .037). No significant associations were observed between the polymorphism with other risk factors for CVD in PW. CONCLUSION In postmenopausal women, the ATG16L1 (T300A) polymorphism is significantly associated with increased carotid IMT (marker of atherosclerotic disease) after adjustments of the confounding variables (P < .037). Thus, identifying the ATG16L1 polymorphism is an important strategy for screening asymptomatic PW who are more predisposed to developing CVD.
Collapse
Affiliation(s)
- Claudio Lera Orsatti
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil.
| | - Marcone Lima Sobreira
- Department of Surgery, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Valéria Cristina Sandrim
- Department of Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Jorge Nahas-Neto
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Fabio Lera Orsatti
- Department of Sport Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Eliana Aguiar Petri Nahas
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
23
|
Abstract
Inflammatory bowel disease [IBD], including ulcerative colitis and Crohn's disease, is a chronic and unpredictable condition characterised by alternating periods of remission interspersed with relapses. In recent years, accumulating support for an immunomodulating effect of vitamin D on both the innate and the adaptive immune systems has been presented. Through the vitamin D receptor, the active form of vitamin D, 1,25[OH]2D, induces antimicrobial peptide secretion, decreases dendritic cell activity, and promotes Th2 and regulatory T cell development and activity. In addition, vitamin D promotes an increased ratio of anti-inflammatory cytokines to pro-inflammatory cytokines. Studies in IBD point to a role for vitamin D in ameliorating disease outcome. Suboptimal circulating levels of 25-hydroxyvitamin D are common in IBD and appear to be associated with an increased risk of flares, IBD-related hospitalisations and surgeries, an inadequate response to tumour necrosis factor [TNF] inhibitors, a deterioration in quality of life, and low bone mineral density. With only few available randomised double-blind, placebo-controlled studies investigating therapeutic effects of vitamin D related to IBD, further research is necessary to determine the true therapeutic potential of vitamin D, as well as to define its optimal range in serum to achieve and maintain quiescence of disease. This review aims to summarise the latest knowledge on the extraskeletal effects of vitamin D in IBD, and outlines the potential deleterious consequences of vitamin D deficiency in this patient cohort.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lars Rejnmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Alan C Moss
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA
| |
Collapse
|
24
|
He S, Li Q, Jiang X, Lu X, Feng F, Qu W, Chen Y, Sun H. Design of Small Molecule Autophagy Modulators: A Promising Druggable Strategy. J Med Chem 2017; 61:4656-4687. [PMID: 29211480 DOI: 10.1021/acs.jmedchem.7b01019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autophagy is a lysosome-dependent mechanism of intracellular degradation for maintaining cellular homeostasis. Dysregulation of autophagy has been verified to be closely linked to a number of human diseases. Consequently, targeting autophagy has been highlighted as a novel therapeutic strategy for clinical utility. Mounting efforts have been done in recent years to elucidate the mechanisms of autophagy regulation and to identify potential modulators of autophagy. However, most of the compounds target complex and multifaceted pathway and proteins, which may limit the evaluation of therapeutic value and in depth studies as chemical tools. Therefore, the development of specific and active autophagy modulators becomes most desirable. Here, we briefly review the regulation of autophagy and then summarize the recent development of small molecules targeting the core autophagic machinery. Finally, we put forward our viewpoints on the current problems, with the aim to provide reference for future drug discovery and potential therapeutic perspectives on novel, potent, selective autophagy modulators.
Collapse
Affiliation(s)
- Siyu He
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| | - Qi Li
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| | - Xueyang Jiang
- Key Laboratory of Biomedical Functional Materials, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| | - Xin Lu
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| | - Feng Feng
- Key Laboratory of Biomedical Functional Materials, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| | - Wei Qu
- Key Laboratory of Biomedical Functional Materials, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| | - Yao Chen
- School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing , 210023 , China
| | - Haopeng Sun
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| |
Collapse
|
25
|
Takahama M, Akira S, Saitoh T. Autophagy limits activation of the inflammasomes. Immunol Rev 2017; 281:62-73. [DOI: 10.1111/imr.12613] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Michihiro Takahama
- Division of Inflammation Biology; Institute for Enzyme Research; Tokushima University; Tokushima Japan
| | - Shizuo Akira
- Laboratory of Host Defense; World Premier International Research Center Immunology Frontier Research Center; Osaka University; Osaka Japan
- Department of Host Defense; Research Institute for Microbial Diseases; Osaka University; Osaka Japan
| | - Tatsuya Saitoh
- Division of Inflammation Biology; Institute for Enzyme Research; Tokushima University; Tokushima Japan
| |
Collapse
|
26
|
Tesselaar MH, Crezee T, Schuurmans I, Gerrits D, Nagarajah J, Boerman OC, van Engen-van Grunsven I, Smit JWA, Netea-Maier RT, Plantinga TS. Digitalislike Compounds Restore hNIS Expression and Iodide Uptake Capacity in Anaplastic Thyroid Cancer. J Nucl Med 2017; 59:780-786. [PMID: 29242405 DOI: 10.2967/jnumed.117.200675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare malignancy that accounts for 1%-2% of all thyroid cancers. ATC is one of the most aggressive human cancers, with rapid growth, tumor invasion, and development of distant metastases. The median survival is only 5 mo, and the 1-y survival is less than 20%. Moreover, as a result of severe dedifferentiation, including the loss of human sodium iodide symporter (hNIS) expression, radioactive iodide (RAI) therapy is ineffective. Recently, we have demonstrated beneficial effects of autophagy-activating digitalislike compounds (DLCs) on redifferentiation and concomitant restoration of iodide uptake in RAI-refractory papillary and follicular thyroid cancer cell lines. In the current study, the effects of DLCs on differentiation and proliferation of ATC cell lines were investigated. Methods: Autophagy activity was assessed in ATC patient tissues by immunofluorescent staining for the autophagy marker microtubule-associated protein 1A/1B-light chain 3 (LC3). In addition, the effect of autophagy-activating DLCs on the proliferation, gene expression profile, and iodide uptake capacity of ATC cell lines was studied. Results: Diminished autophagy activity was observed in ATC tissues, and in vitro treatment of ATC cell lines with DLCs robustly restored hNIS and thyroglobulin expression and iodide uptake capacity. In addition, proliferation was strongly reduced by induction of cell cycle arrest and, to some extent, cell death. Mechanistically, reactivation of functional hNIS expression could be attributed to activation of the transcription factors activating transcription factor 3 and protooncogene c-fosConclusion: DLCs could represent a promising adjunctive therapy for restoring iodide avidity within the full spectrum from RAI-refractory dedifferentiated to ATC.
Collapse
Affiliation(s)
- Marika H Tesselaar
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Thomas Crezee
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Imke Schuurmans
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Danny Gerrits
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands; and
| | - James Nagarajah
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands; and
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands; and
| | - Ilse van Engen-van Grunsven
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johannes W A Smit
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Theo S Plantinga
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Schwerd T, Pandey S, Yang HT, Bagola K, Jameson E, Jung J, Lachmann RH, Shah N, Patel SY, Booth C, Runz H, Düker G, Bettels R, Rohrbach M, Kugathasan S, Chapel H, Keshav S, Elkadri A, Platt N, Muise AM, Koletzko S, Xavier RJ, Marquardt T, Powrie F, Wraith JE, Gyrd-Hansen M, Platt FM, Uhlig HH. Impaired antibacterial autophagy links granulomatous intestinal inflammation in Niemann-Pick disease type C1 and XIAP deficiency with NOD2 variants in Crohn's disease. Gut 2017; 66:1060-1073. [PMID: 26953272 PMCID: PMC5532464 DOI: 10.1136/gutjnl-2015-310382] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 01/06/2016] [Accepted: 01/14/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Patients with Niemann-Pick disease type C1 (NPC1), a lysosomal lipid storage disorder that causes neurodegeneration and liver damage, can present with IBD, but neither the significance nor the functional mechanism of this association is clear. We studied bacterial handling and antibacterial autophagy in patients with NPC1. DESIGN We characterised intestinal inflammation in 14 patients with NPC1 who developed IBD. We investigated bacterial handling and cytokine production of NPC1 monocytes or macrophages in vitro and compared NPC1-associated functional defects to those caused by IBD-associated nucleotide-binding oligomerization domain-containing protein 2 (NOD2) variants or mutations in X-linked inhibitor of apoptosis (XIAP). RESULTS Patients with the lysosomal lipid storage disorder NPC1 have increased susceptibility to early-onset fistulising colitis with granuloma formation, reminiscent of Crohn's disease (CD). Mutations in NPC1 cause impaired autophagy due to defective autophagosome function that abolishes NOD2-mediated bacterial handling in vitro similar to variants in NOD2 or XIAP deficiency. In contrast to genetic NOD2 and XIAP variants, NPC1 mutations do not impair NOD2-receptor-interacting kinase 2 (RIPK2)-XIAP-dependent cytokine production. Pharmacological activation of autophagy can rescue bacterial clearance in macrophages in vitro by increasing the autophagic flux and bypassing defects in NPC1. CONCLUSIONS NPC1 confers increased risk of early-onset severe CD. Our data support the concept that genetic defects at different checkpoints of selective autophagy cause a shared outcome of CD-like immunopathology linking monogenic and polygenic forms of IBD. Muramyl dipeptide-driven cytokine responses and antibacterial autophagy induction are parallel and independent signalling cascades downstream of the NOD2-RIPK2-XIAP complex.
Collapse
Affiliation(s)
- Tobias Schwerd
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sumeet Pandey
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Huei-Ting Yang
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Katrin Bagola
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Elisabeth Jameson
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| | - Jonathan Jung
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | | | - Neil Shah
- Great Ormond Street Hospital, London, UK
| | - Smita Y Patel
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Claire Booth
- Department of Clinical Immunology, Great Ormond Street Hospital, London, UK
| | - Heiko Runz
- University of Heidelberg, Heidelberg, Germany
| | - Gesche Düker
- University Children's Hospital Bonn, Bonn, Germany
| | | | - Marianne Rohrbach
- Children's Research Centre Zurich, University Children's Hospital, Zurich, Switzerland
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Helen Chapel
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Satish Keshav
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Abdul Elkadri
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada,Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Nick Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Alexio M Muise
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada,Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Ramnik J Xavier
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Fiona Powrie
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK,Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - James E Wraith
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| | - Mads Gyrd-Hansen
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK,Department of Pediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
28
|
Uniken Venema WT, Voskuil MD, Dijkstra G, Weersma RK, Festen EA. The genetic background of inflammatory bowel disease: from correlation to causality. J Pathol 2016; 241:146-158. [PMID: 27785786 DOI: 10.1002/path.4817] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/13/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022]
Abstract
Recent studies have greatly improved our insight into the genetic background of inflammatory bowel disease (IBD). New high-throughput technologies and large-scale international collaborations have contributed to the identification of 200 independent genetic risk loci for IBD. However, in most of these loci, it is unclear which gene conveys the risk for IBD. More importantly, it is unclear which variant within or near the gene is causal to the disease. Using targeted GWAS, imputation, resequencing of risk loci, and in silico fine-mapping of densely typed loci, several causal variants have been identified in IBD risk genes, and various pathological pathways have been uncovered. Current research in the field of IBD focuses on the effect of these causal variants on gene expression and protein function. However, more elements than only the genome must be taken into account to disentangle the multifactorial pathology of IBD. The genetic risk loci identified to date only explain a small part of genetic variance in disease risk. Currently, large multi-omics studies are incorporating factors ranging from the gut microbiome to the environment. In this review, we present the progress that has been made in IBD genetic research and stress the importance of studying causality to increase our understanding of the pathogenesis of IBD. We highlight important causal genetic variants in the candidate genes NOD2, ATG16L1, IRGM, IL23R, CARD9, RNF186, and PRDM1. We describe their downstream effects on protein function and their direct effects on the gut immune system. Furthermore, we discuss the future role of genetics in unravelling disease mechanisms in IBD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Werna Tc Uniken Venema
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel D Voskuil
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Eleonora Am Festen
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.,Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Tesselaar MH, Crezee T, Swarts HG, Gerrits D, Boerman OC, Koenderink JB, Stunnenberg HG, Netea MG, Smit JW, Netea-Maier RT, Plantinga TS. Digitalis-like Compounds Facilitate Non-Medullary Thyroid Cancer Redifferentiation through Intracellular Ca2+, FOS, and Autophagy-Dependent Pathways. Mol Cancer Ther 2016; 16:169-181. [DOI: 10.1158/1535-7163.mct-16-0460] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/26/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
|
30
|
Fan Y, Wang N, Rocchi A, Zhang W, Vassar R, Zhou Y, He C. Identification of natural products with neuronal and metabolic benefits through autophagy induction. Autophagy 2016; 13:41-56. [PMID: 27791467 DOI: 10.1080/15548627.2016.1240855] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a housekeeping lysosomal degradation pathway important for cellular survival, homeostasis and function. Various disease models have shown that upregulation of autophagy may be beneficial to combat disease pathogenesis. However, despite several recently reported small-molecule screens for synthetic autophagy inducers, natural chemicals of diverse structures and functions have not been included in the synthetic libraries, and characterization of their roles in autophagy has been lacking. To discover novel autophagy-regulating compounds and study their therapeutic mechanisms, we used analytic chemistry approaches to isolate natural phytochemicals from a reservoir of medicinal plants used in traditional remedies. From this pilot plant metabolite library, we identified several novel autophagy-inducing phytochemicals, including Rg2. Rg2 is a steroid glycoside chemical that activates autophagy in an AMPK-ULK1-dependent and MTOR-independent manner. Induction of autophagy by Rg2 enhances the clearance of protein aggregates in a cell-based model, improves cognitive behaviors in a mouse model of Alzheimer disease, and prevents high-fat diet-induced insulin resistance. Thus, we discovered a series of autophagy-inducing phytochemicals from medicinal plants, and found that one of the compounds Rg2 mediates metabolic and neurotrophic effects dependent on activation of the autophagy pathway. These findings may help explain how medicinal plants exert the therapeutic functions against metabolic diseases.
Collapse
Affiliation(s)
- Yuying Fan
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA.,b Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University , Changchun, Jilin , China
| | - Nan Wang
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA.,c Key Laboratory of Industrial Microbiology , Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology , Tianjin , China
| | - Altea Rocchi
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Weiran Zhang
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Robert Vassar
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Yifa Zhou
- b Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University , Changchun, Jilin , China
| | - Congcong He
- a Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| |
Collapse
|
31
|
Abstract
Historically, the brain has been considered an immune-privileged organ separated from the peripheral immune system by the blood-brain barrier. However, immune responses do occur in the brain in neurological conditions in which the integrity of the blood-brain barrier is compromised, exposing the brain to peripheral antigens and endogenous danger signals. While most of the associated pathological processes occur in the central nervous system, it is now clear that peripheral immune cells, especially mononuclear phagocytes, that infiltrate into the injury site play a key role in modulating the progression of primary brain injury development. As inflammation is a necessary and critical component for the subsequent injury resolution process, understanding the contribution of mononuclear phagocytes on the regulation of inflammatory responses may provide novel approaches for potential therapies. Furthermore, predisposed comorbid conditions at the time of stroke cause the alteration of stroke-induced immune and inflammatory responses and subsequently influence stroke outcome. In this review, we summarize a role for microglia and monocytes/macrophages in acute ischemic stroke in the context of normal and metabolically compromised conditions.
Collapse
Affiliation(s)
- Eunhee Kim
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Medical Research Institute, White Plains, NY, 10605, USA
| | - Sunghee Cho
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Medical Research Institute, White Plains, NY, 10605, USA.
| |
Collapse
|
32
|
Natural Compounds as Regulators of NLRP3 Inflammasome-Mediated IL-1 β Production. Mediators Inflamm 2016; 2016:5460302. [PMID: 27672241 PMCID: PMC5031844 DOI: 10.1155/2016/5460302] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/18/2016] [Accepted: 08/11/2016] [Indexed: 12/17/2022] Open
Abstract
IL-1β is one of the main proinflammatory cytokines that regulates a broad range of immune responses and also participates in several physiological processes. The canonical production of IL-1β requires multiprotein complexes called inflammasomes. One of the most intensively studied inflammasome complexes is the NLRP3 inflammasome. Its activation requires two signals: one signal “primes” the cells and induces the expression of NLRP3 and pro-IL-1β, while the other signal leads to the assembly and activation of the complex. Several stimuli were reported to function as the second signal including reactive oxygen species, lysosomal rupture, or cytosolic ion perturbation. Despite very intensive studies, the precise function and regulation of the NLRP3 inflammasome are still not clear. However, many chronic inflammatory diseases are related to the overproduction of IL-1β that is mediated via the NLRP3 inflammasome. In this review, we aimed to provide an overview of studies that demonstrated the effect of plant-derived natural compounds on NLRP3 inflammasome-mediated IL-1β production. Although many of these studies lack the mechanistic explanation of their action, these compounds may be considered as complementary supplements in the treatment of chronic inflammatory diseases, consumed as preventive agents, and may also be considered as molecular tools to study NLRP3 function.
Collapse
|
33
|
Salzer HJF, Wassilew N, Köhler N, Olaru ID, Günther G, Herzmann C, Kalsdorf B, Sanchez-Carballo P, Terhalle E, Rolling T, Lange C, Heyckendorf J. Personalized Medicine for Chronic Respiratory Infectious Diseases: Tuberculosis, Nontuberculous Mycobacterial Pulmonary Diseases, and Chronic Pulmonary Aspergillosis. Respiration 2016; 92:199-214. [PMID: 27595540 DOI: 10.1159/000449037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chronic respiratory infectious diseases are causing high rates of morbidity and mortality worldwide. Tuberculosis, a major cause of chronic pulmonary infection, is currently responsible for approximately 1.5 million deaths per year. Although important advances in the fight against tuberculosis have been made, the progress towards eradication of this disease is being challenged by the dramatic increase in multidrug-resistant bacilli. Nontuberculous mycobacteria causing pulmonary disease and chronic pulmonary aspergillosis are emerging infectious diseases. In contrast to other infectious diseases, chronic respiratory infections share the trait of having highly variable treatment outcomes despite longstanding antimicrobial therapy. Recent scientific progress indicates that medicine is presently at a transition stage from programmatic to personalized management. We explain current state-of-the-art management concepts of chronic pulmonary infectious diseases as well as the underlying methods for therapeutic decisions and their implications for personalized medicine. Furthermore, we describe promising biomarkers and techniques with the potential to serve future individual treatment concepts in this field of difficult-to-treat patients. These include candidate markers to improve individual risk assessment for disease development, the design of tailor-made drug therapy regimens, and individualized biomarker-guided therapy duration to achieve relapse-free cure. In addition, the use of therapeutic drug monitoring to reach optimal drug dosing with the smallest rate of adverse events as well as candidate agents for future host-directed therapies are described. Taken together, personalized medicine will provide opportunities to substantially improve the management and treatment outcome of difficult-to-treat patients with chronic respiratory infections.
Collapse
Affiliation(s)
- Helmut J F Salzer
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gupta A, Misra A, Deretic V. Targeted pulmonary delivery of inducers of host macrophage autophagy as a potential host-directed chemotherapy of tuberculosis. Adv Drug Deliv Rev 2016; 102:10-20. [PMID: 26829287 DOI: 10.1016/j.addr.2016.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/26/2015] [Accepted: 01/21/2016] [Indexed: 12/19/2022]
Abstract
One of the promising host-directed chemotherapeutic interventions in tuberculosis (TB) is based on inducing autophagy as an immune effector. Here we consider the strengths and weaknesses of potential autophagy-based pharmacological intervention. Using the existing drugs that induce autophagy is an option, but it has limitations given the broad role of autophagy in most cells, tissues, and organs. Thus, it may be desirable that the agent being used to modulate autophagy is applied in a targeted manner, e.g. delivered to affected tissues, with infected macrophages being an obvious choice. This review addresses the advantages and disadvantages of delivering drugs to induce autophagy in M. tuberculosis-infected macrophages. One option, already being tested in models, is to design particles for inhalation delivery to lung macrophages. The choice of drugs, drug release kinetics and intracellular residence times, non-target cell exposure and feasibility of use by patients is discussed. We term here this (still experimental) approach, of compartment-targeting, autophagy-based, host-directed therapy as "Track-II antituberculosis chemotherapy."
Collapse
|
35
|
Kabat AM, Pott J, Maloy KJ. The Mucosal Immune System and Its Regulation by Autophagy. Front Immunol 2016; 7:240. [PMID: 27446072 PMCID: PMC4916208 DOI: 10.3389/fimmu.2016.00240] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 06/07/2016] [Indexed: 12/20/2022] Open
Abstract
The gastrointestinal tract presents a unique challenge to the mucosal immune system, which has to constantly monitor the vast surface for the presence of pathogens, while at the same time maintaining tolerance to beneficial or innocuous antigens. In the intestinal mucosa, specialized innate and adaptive immune components participate in directing appropriate immune responses toward these diverse challenges. Recent studies provide compelling evidence that the process of autophagy influences several aspects of mucosal immune responses. Initially described as a “self-eating” survival pathway that enables nutrient recycling during starvation, autophagy has now been connected to multiple cellular responses, including several aspects of immunity. Initial links between autophagy and host immunity came from the observations that autophagy can target intracellular bacteria for degradation. However, subsequent studies indicated that autophagy plays a much broader role in immune responses, as it can impact antigen processing, thymic selection, lymphocyte homeostasis, and the regulation of immunoglobulin and cytokine secretion. In this review, we provide a comprehensive overview of mucosal immune cells and discuss how autophagy influences many aspects of their physiology and function. We focus on cell type-specific roles of autophagy in the gut, with a particular emphasis on the effects of autophagy on the intestinal T cell compartment. We also provide a perspective on how manipulation of autophagy may potentially be used to treat mucosal inflammatory disorders.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| |
Collapse
|
36
|
Lassen KG, McKenzie CI, Mari M, Murano T, Begun J, Baxt LA, Goel G, Villablanca EJ, Kuo SY, Huang H, Macia L, Bhan AK, Batten M, Daly MJ, Reggiori F, Mackay CR, Xavier RJ. Genetic Coding Variant in GPR65 Alters Lysosomal pH and Links Lysosomal Dysfunction with Colitis Risk. Immunity 2016; 44:1392-405. [PMID: 27287411 DOI: 10.1016/j.immuni.2016.05.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/19/2016] [Accepted: 03/21/2016] [Indexed: 12/28/2022]
Abstract
Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.
Collapse
Affiliation(s)
- Kara G Lassen
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Craig I McKenzie
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Muriel Mari
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, 3713 AV Groningen, the Netherlands; Department of Cell Biology, University Medical Center Utrecht, 3564 CX Utrecht, the Netherlands
| | - Tatsuro Murano
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jakob Begun
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Mater Research Institute and School of Medicine, University of Queensland, Brisbane, QLD 4101, Australia
| | - Leigh A Baxt
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gautam Goel
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eduardo J Villablanca
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Szu-Yu Kuo
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hailiang Huang
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Laurence Macia
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Atul K Bhan
- Pathology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marcel Batten
- Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Mark J Daly
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fulvio Reggiori
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, 3713 AV Groningen, the Netherlands; Department of Cell Biology, University Medical Center Utrecht, 3564 CX Utrecht, the Netherlands
| | - Charles R Mackay
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
37
|
Saitoh T, Akira S. Regulation of inflammasomes by autophagy. J Allergy Clin Immunol 2016; 138:28-36. [PMID: 27373323 DOI: 10.1016/j.jaci.2016.05.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022]
Abstract
Inflammasomes detect pathogen-associated molecular patterns to induce inflammatory innate immune responses and play a key role in host defense against infectious agents. However, inflammasomes are often wrongly activated by metabolites, amyloids, and environmental irritants. This induces massive inflammation, causing severe tissue damage, and results in the development of inflammatory diseases. Hence cellular machineries regulating both "activation" and "inactivation" of inflammasomes are definitely important. Recent studies have shown that autophagy, an intracellular degradation system associated with maintenance of cellular homeostasis, plays a key role in inflammasome inactivation. Notably, autophagy deficiency caused by gene mutation disrupts organelle elimination and thus induces aberrant activation of inflammasomes, leading to severe tissue damage. Here we review recent findings regarding the involvement of autophagy in the regulation of inflammasome activation and development of inflammatory disorders.
Collapse
Affiliation(s)
- Tatsuya Saitoh
- Department of Inflammation Biology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Research Center Immunology Frontier Research Center, Osaka, Japan; Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
38
|
Yang S, Imamura Y, Jenkins RW, Cañadas I, Kitajima S, Aref A, Brannon A, Oki E, Castoreno A, Zhu Z, Thai T, Reibel J, Qian Z, Ogino S, Wong KK, Baba H, Kimmelman AC, Pasca Di Magliano M, Barbie DA. Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation. Cancer Immunol Res 2016; 4:520-30. [PMID: 27068336 DOI: 10.1158/2326-6066.cir-15-0235] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
Autophagy promotes tumor progression downstream of oncogenic KRAS, yet also restrains inflammation and dysplasia through mechanisms that remain incompletely characterized. Understanding the basis of this paradox has important implications for the optimal targeting of autophagy in cancer. Using a mouse model of cerulein-induced pancreatitis, we found that loss of autophagy by deletion of Atg5 enhanced activation of the IκB kinase (IKK)-related kinase TBK1 in vivo, associated with increased neutrophil and T-cell infiltration and PD-L1 upregulation. Consistent with this observation, pharmacologic or genetic inhibition of autophagy in pancreatic ductal adenocarcinoma cells, including suppression of the autophagy receptors NDP52 or p62, prolonged TBK1 activation and increased expression of CCL5, IL6, and several other T-cell and neutrophil chemotactic cytokines in vitro Defective autophagy also promoted PD-L1 upregulation, which is particularly pronounced downstream of IFNγ signaling and involves JAK pathway activation. Treatment with the TBK1/IKKε/JAK inhibitor CYT387 (also known as momelotinib) not only inhibits autophagy, but also suppresses this feedback inflammation and reduces PD-L1 expression, limiting KRAS-driven pancreatic dysplasia. These findings could contribute to the dual role of autophagy in oncogenesis and have important consequences for its therapeutic targeting. Cancer Immunol Res; 4(6); 520-30. ©2016 AACR.
Collapse
Affiliation(s)
- Shenghong Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Yu Imamura
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan. Department of Gastroenterological Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan. Department of Surgery and Science, Graduate of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Russell W Jenkins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Israel Cañadas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Shunsuke Kitajima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Amir Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Arthur Brannon
- Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan. Cell and Developmental Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Eiji Oki
- Department of Surgery and Science, Graduate of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Adam Castoreno
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Zehua Zhu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Tran Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Jacob Reibel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhirong Qian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Kwok K Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Alec C Kimmelman
- Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marina Pasca Di Magliano
- Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan. Cell and Developmental Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
| |
Collapse
|
39
|
Identification of a novel interaction between corticotropin releasing hormone (Crh) and macroautophagy. Sci Rep 2016; 6:23342. [PMID: 26987580 PMCID: PMC4796911 DOI: 10.1038/srep23342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 03/02/2016] [Indexed: 02/07/2023] Open
Abstract
In inflammatory bowel disease (IBD), compromised restitution of the epithelial barrier contributes to disease severity. Owing to the complexity in the pathogenesis of IBD, a variety of factors have been implicated in its progress. In this study, we report a functional interaction between macroautophagy and Corticotropin Releasing Hormone (Crh) in the gut. For this purpose we used DSS colitis model on Crh -/- or wild-type (wt) with pharmacological inhibition of autophagy. We uncovered sustained basal autophagy in the gut of Crh -/- mice, which persisted over the course of DSS administration. Autophagy inhibition resulted in partial rescue of Crh -/- mice, while it increased the expression of Crh in the wt gut. Similarly, Crh deficiency was associated with sustained activation of base line autophagy. In vitro models of amino acid deprivation- and LPS-induced autophagy confirmed the in vivo findings. Our results indicate a novel role for Crh in the intestinal epithelium that involves regulation of autophagy, while suggesting the complementary action of the two pathways. These data suggest the intriguing possibility that targeting Crh stimulation in the intestine may provide a novel therapeutic approach to support the integrity of the epithelial barrier and to protect from chronic colitis.
Collapse
|
40
|
Kabat AM, Harrison OJ, Riffelmacher T, Moghaddam AE, Pearson CF, Laing A, Abeler-Dörner L, Forman SP, Grencis RK, Sattentau Q, Simon AK, Pott J, Maloy KJ. The autophagy gene Atg16l1 differentially regulates Treg and TH2 cells to control intestinal inflammation. eLife 2016; 5:e12444. [PMID: 26910010 PMCID: PMC4798959 DOI: 10.7554/elife.12444] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/23/2016] [Indexed: 12/19/2022] Open
Abstract
A polymorphism in the autophagy gene Atg16l1 is associated with susceptibility to inflammatory bowel disease (IBD); however, it remains unclear how autophagy contributes to intestinal immune homeostasis. Here, we demonstrate that autophagy is essential for maintenance of balanced CD4+ T cell responses in the intestine. Selective deletion of Atg16l1 in T cells in mice resulted in spontaneous intestinal inflammation that was characterized by aberrant type 2 responses to dietary and microbiota antigens, and by a loss of Foxp3+ Treg cells. Specific ablation of Atg16l1 in Foxp3+ Treg cells in mice demonstrated that autophagy directly promotes their survival and metabolic adaptation in the intestine. Moreover, we also identify an unexpected role for autophagy in directly limiting mucosal TH2 cell expansion. These findings provide new insights into the reciprocal control of distinct intestinal TH cell responses by autophagy, with important implications for understanding and treatment of chronic inflammatory disorders. DOI:http://dx.doi.org/10.7554/eLife.12444.001 The gut presents a puzzle to our immune system. Immune cells must rapidly respond to antigens produced by harmful bacteria, but food and the beneficial bacteria that inhabit the gut also produce antigens that our immune system must tolerate. Inappropriate immune responses in the gut can lead to inflammatory bowel disease, a debilitating disease with no current cure. We do not fully understand why these harmful inflammatory responses arise, but we know that genetic factors are important. Mutations in genes that affect a process known as autophagy – a pathway that breaks down and recycles unwanted material inside cells – make inflammatory bowel disease more likely to develop, but exactly how they do so remains unclear. T helper cells are crucial controllers of intestinal immune responses and changes in their numbers and behaviour occur during inflammatory bowel disease. Kabat et al. explored how the autophagy pathway affects these key immune cells in mice. Blocking autophagy in T cells altered the balance of different types of T helper cells in the gut. A crucial population of regulatory T cells, which keep inflammatory responses in check, was lost. At the same time, another population of T cells expanded: the T helper 2 (TH2) cells that are responsible for driving allergies. As a result, the mice developed intestinal inflammation and produced antibodies against gut bacteria and food. Overall, Kabat et al.’s results show that autophagy defects can alter the balance of different types of T cells in the gut, leading to inflammation in the intestine. These observations contribute to our understanding of how genetic changes may influence susceptibility to inflammatory bowel disease. They also suggest that drugs that activate autophagy could help to treat diseases associated with changes in regulatory T cells or TH2 cells, including inflammatory bowel disease and allergies. It will now be important to test this and to confirm whether similar changes in T cells are present in humans that have mutations in autophagy genes. DOI:http://dx.doi.org/10.7554/eLife.12444.002
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Oliver J Harrison
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.,Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Thomas Riffelmacher
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Amin E Moghaddam
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Claire F Pearson
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Adam Laing
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Lucie Abeler-Dörner
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Simon P Forman
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - Richard K Grencis
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - Quentin Sattentau
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Anna Katharina Simon
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Chauhan S, Ahmed Z, Bradfute SB, Arko-Mensah J, Mandell MA, Won Choi S, Kimura T, Blanchet F, Waller A, Mudd MH, Jiang S, Sklar L, Timmins GS, Maphis N, Bhaskar K, Piguet V, Deretic V. Pharmaceutical screen identifies novel target processes for activation of autophagy with a broad translational potential. Nat Commun 2015; 6:8620. [PMID: 26503418 PMCID: PMC4624223 DOI: 10.1038/ncomms9620] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a conserved homeostatic process active in all human cells and affecting a spectrum of diseases. Here we use a pharmaceutical screen to discover new mechanisms for activation of autophagy. We identify a subset of pharmaceuticals inducing autophagic flux with effects in diverse cellular systems modelling specific stages of several human diseases such as HIV transmission and hyperphosphorylated tau accumulation in Alzheimer's disease. One drug, flubendazole, is a potent inducer of autophagy initiation and flux by affecting acetylated and dynamic microtubules in a reciprocal way. Disruption of dynamic microtubules by flubendazole results in mTOR deactivation and dissociation from lysosomes leading to TFEB (transcription factor EB) nuclear translocation and activation of autophagy. By inducing microtubule acetylation, flubendazole activates JNK1 leading to Bcl-2 phosphorylation, causing release of Beclin1 from Bcl-2-Beclin1 complexes for autophagy induction, thus uncovering a new approach to inducing autophagic flux that may be applicable in disease treatment. Autophagy is a homeostatic process that could be a potential drug target in the treatment of disease. Here the authors identify in a pharmaceutical screen flubendazole as an inducer of autophagy initiation and flux by affecting microtubules, mTOR, TFEB and Beclin 1 activity.
Collapse
Affiliation(s)
- Santosh Chauhan
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Zahra Ahmed
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Steven B Bradfute
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - John Arko-Mensah
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michael A Mandell
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Seong Won Choi
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Tomonori Kimura
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Fabien Blanchet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Anna Waller
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michal H Mudd
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Shanya Jiang
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Larry Sklar
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Graham S Timmins
- College of Pharmacy, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Nicole Maphis
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Vincent Piguet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
42
|
Sadaghian Sadabad M, Regeling A, de Goffau MC, Blokzijl T, Weersma RK, Penders J, Faber KN, Harmsen HJM, Dijkstra G. The ATG16L1-T300A allele impairs clearance of pathosymbionts in the inflamed ileal mucosa of Crohn's disease patients. Gut 2015; 64:1546-52. [PMID: 25253126 DOI: 10.1136/gutjnl-2014-307289] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 09/09/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Crohn's disease (CD) is caused by a complex interplay among genetic, microbial and environmental factors. ATG16L1 is an important genetic factor involved in innate immunity, including autophagy and phagocytosis of microbial components from the gut. We investigated the effect of inflammation on the composition of microbiota in the ileal mucosa of CD patients in relation to the ATG16L1 risk status. DESIGN Biopsies (n=35) were obtained from inflamed and non-inflamed regions of the terminal ileum of 11 CD patients homozygous for the ATG16L1 risk allele (ATG16L1-T300A) and 9 CD patients homozygous for the ATG16L1 protective allele (ATG16L1-T300). Biopsy DNA was extracted and the bacterial composition analysed by pyrosequencing. Intracellular survival rates of adherent-invasive Escherichia coli (AIEC) were analysed by determining colony forming units after exposure to monocytes isolated from healthy volunteers homozygous for the ATG16L1 risk or protective allele. RESULTS Inflamed ileal tissue from patients homozygous for the ATG16L1 risk allele contained increased numbers of Fusobacteriaceae, whereas inflamed ileal tissue of patients homozygous for the ATG16L1 protective allele showed decreased numbers of Bacteroidaceae and Enterobacteriaceae and increased Lachnospiraceae. The ATG16L1 allele did not affect the bacterial composition in the non-inflamed ileal tissue. Monocytes homozygous for the ATG16L1 risk allele showed impaired killing of AIEC under inflammatory conditions compared with those homozygous for the ATG16L1 protective allele. CONCLUSIONS CD patients homozygous for the ATG16L1-T300A risk allele show impaired clearance of pathosymbionts in ileal inflammation indicating that ATG16L1 is essential for effective elimination of pathosymbionts upon inflammation.
Collapse
Affiliation(s)
- Mehdi Sadaghian Sadabad
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anouk Regeling
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcus C de Goffau
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tjasso Blokzijl
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Penders
- Department of Medical Microbiology, School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
43
|
Autophagy and autophagy-related proteins in the immune system. Nat Immunol 2015; 16:1014-24. [DOI: 10.1038/ni.3273] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/17/2015] [Indexed: 02/06/2023]
|
44
|
Baxt LA, Xavier RJ. Role of Autophagy in the Maintenance of Intestinal Homeostasis. Gastroenterology 2015; 149:553-62. [PMID: 26170139 PMCID: PMC4550567 DOI: 10.1053/j.gastro.2015.06.046] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/16/2015] [Accepted: 06/29/2015] [Indexed: 02/08/2023]
Abstract
Genome-wide association studies of inflammatory bowel disease have identified several risk loci in genes that regulate autophagy, and studies have provided insight into the functional effects of these polymorphisms. We review the mechanisms by which autophagy contributes to intestinal homeostasis, focusing on its cell type-specific roles in regulating gut ecology, restricting pathogenic bacteria, and controlling inflammation. Based on this information, we are beginning to understand how alterations in autophagy can contribute to intestinal inflammation.
Collapse
Affiliation(s)
- Leigh A. Baxt
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts,Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Interactions between Autophagy and Bacterial Toxins: Targets for Therapy? Toxins (Basel) 2015; 7:2918-58. [PMID: 26248079 PMCID: PMC4549733 DOI: 10.3390/toxins7082918] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 01/07/2023] Open
Abstract
Autophagy is a physiological process involved in defense mechanisms for clearing intracellular bacteria. The autophagic pathway is finely regulated and bacterial toxins interact with this process in a complex manner. Bacterial toxins also interact significantly with many biochemical processes. Evaluations of the effects of bacterial toxins, such as endotoxins, pore-forming toxins and adenylate cyclases, on autophagy could support the development of new strategies for counteracting bacterial pathogenicity. Treatment strategies could focus on drugs that enhance autophagic processes to improve the clearance of intracellular bacteria. However, further in vivo studies are required to decipher the upregulation of autophagy and potential side effects limiting such approaches. The capacity of autophagy activation strategies to improve the outcome of antibiotic treatment should be investigated in the future.
Collapse
|
46
|
Small-molecule enhancers of autophagy modulate cellular disease phenotypes suggested by human genetics. Proc Natl Acad Sci U S A 2015. [PMID: 26195741 DOI: 10.1073/pnas.1512289112] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Studies of human genetics and pathophysiology have implicated the regulation of autophagy in inflammation, neurodegeneration, infection, and autoimmunity. These findings have motivated the use of small-molecule probes to study how modulation of autophagy affects disease-associated phenotypes. Here, we describe the discovery of the small-molecule probe BRD5631 that is derived from diversity-oriented synthesis and enhances autophagy through an mTOR-independent pathway. We demonstrate that BRD5631 affects several cellular disease phenotypes previously linked to autophagy, including protein aggregation, cell survival, bacterial replication, and inflammatory cytokine production. BRD5631 can serve as a valuable tool for studying the role of autophagy in the context of cellular homeostasis and disease.
Collapse
|
47
|
Khor B, Gagnon JD, Goel G, Roche MI, Conway KL, Tran K, Aldrich LN, Sundberg TB, Paterson AM, Mordecai S, Dombkowski D, Schirmer M, Tan PH, Bhan AK, Roychoudhuri R, Restifo NP, O'Shea JJ, Medoff BD, Shamji AF, Schreiber SL, Sharpe AH, Shaw SY, Xavier RJ. The kinase DYRK1A reciprocally regulates the differentiation of Th17 and regulatory T cells. eLife 2015; 4:e05920. [PMID: 25998054 PMCID: PMC4441007 DOI: 10.7554/elife.05920] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 04/27/2015] [Indexed: 12/12/2022] Open
Abstract
The balance between Th17 and T regulatory (Treg) cells critically modulates immune homeostasis, with an inadequate Treg response contributing to inflammatory disease. Using an unbiased chemical biology approach, we identified a novel role for the dual specificity tyrosine-phosphorylation-regulated kinase DYRK1A in regulating this balance. Inhibition of DYRK1A enhances Treg differentiation and impairs Th17 differentiation without affecting known pathways of Treg/Th17 differentiation. Thus, DYRK1A represents a novel mechanistic node at the branch point between commitment to either Treg or Th17 lineages. Importantly, both Treg cells generated using the DYRK1A inhibitor harmine and direct administration of harmine itself potently attenuate inflammation in multiple experimental models of systemic autoimmunity and mucosal inflammation. Our results identify DYRK1A as a physiologically relevant regulator of Treg cell differentiation and suggest a broader role for other DYRK family members in immune homeostasis. These results are discussed in the context of human diseases associated with dysregulated DYRK activity.
Collapse
Affiliation(s)
- Bernard Khor
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Broad Institute of MIT and Harvard, Cambridge, United States
- Pathology Service, Massachusetts General Hospital, Boston, United States
| | - John D Gagnon
- Broad Institute of MIT and Harvard, Cambridge, United States
| | - Gautam Goel
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Marly I Roche
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, United States
| | - Kara L Conway
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Broad Institute of MIT and Harvard, Cambridge, United States
| | - Khoa Tran
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Leslie N Aldrich
- Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | | | - Alison M Paterson
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, United States
| | - Scott Mordecai
- Pathology Service, Massachusetts General Hospital, Boston, United States
| | - David Dombkowski
- Pathology Service, Massachusetts General Hospital, Boston, United States
| | | | - Pauline H Tan
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Atul K Bhan
- Pathology Service, Massachusetts General Hospital, Boston, United States
| | - Rahul Roychoudhuri
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Benjamin D Medoff
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, United States
| | | | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, United States
| | - Stanley Y Shaw
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
- Broad Institute of MIT and Harvard, Cambridge, United States
| |
Collapse
|
48
|
Abstract
Autophagy is an important intracellular catabolic mechanism critically involved in regulating tissue homeostasis. The implication of autophagy in human diseases and the need to understand its regulatory mechanisms in mammalian cells have stimulated research efforts that led to the development of high-throughput screening protocols and small-molecule modulators that can activate or inhibit autophagy. Herein we review the current landscape in the development of screening technology as well as the molecules and pharmacologic agents targeting the regulatory mechanisms of autophagy. We also evaluate the potential therapeutic application of these compounds in different human pathologies.
Collapse
|
49
|
Applying the logic of genetic interaction to discover small molecules that functionally interact with human disease alleles. Methods Mol Biol 2015; 1263:15-27. [PMID: 25618333 PMCID: PMC4357233 DOI: 10.1007/978-1-4939-2269-7_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Despite rapid advances in the genetics of complex human diseases, understanding the significance of human disease alleles remains a critical roadblock to clinical translation. Here, we present a chemical biology approach that uses perturbation with small molecules of known mechanism to reveal mechanistic and therapeutic consequences of human disease alleles. To maximize human applicability, we perform chemical screening on multiple cell lines isolated from individual patients, allowing the effects of disease alleles to be studied in their native genetic context. Chemical screen analysis combines the logic of traditional genetic interaction screens with analytic methods from high-dimensionality gene expression analyses. We rank compounds according to their ability to discriminate between cell lines that are mutant versus wild type at a disease gene (i.e., the compounds induce phenotypes that differ the most across the two classes). A technique called compound set enrichment analysis (CSEA), modeled after a widely used method to identify pathways from gene expression data, identifies sets of functionally or structurally related compounds that are statistically enriched among the most discriminating compounds. This chemical:genetic interaction approach was applied to patient-derived cells in a monogenic form of diabetes and identified several classes of compounds (including FDA-approved drugs) that show functional interactions with the causative disease gene, and also modulate insulin secretion, a critical disease phenotype. In summary, perturbation of patient-derived cells with small molecules of known mechanism, together with compound-set-based pathway analysis, can identify small molecules and pathways that functionally interact with disease alleles, and that can modulate disease networks for therapeutic effect.
Collapse
|
50
|
Sundberg TB, Xavier RJ, Schreiber SL, Shamji AF. Small-molecule control of cytokine function: new opportunities for treating immune disorders. Curr Opin Chem Biol 2014; 23:23-30. [PMID: 25222143 DOI: 10.1016/j.cbpa.2014.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/15/2014] [Accepted: 08/22/2014] [Indexed: 12/29/2022]
Abstract
Manipulating cytokine function with protein-based drugs has proven effective for treating a wide variety of autoimmune and autoinflammatory disorders. However, the limited ability of protein-based drugs to modulate intracellular targets, including many implicated by studies of the genetics and physiology of these diseases, and to coordinately neutralize redundant inflammatory cytokines, suggests an important and complementary role for small molecules in immunomodulatory drug development. The recent clinical approval of Janus kinase and phosphodiesterase inhibitors, along with emerging evidence from other compound classes, firmly establish small molecules as effective tools for modulating therapeutically relevant proteins that give rise to aberrant cytokine signaling or mediate its downstream consequences.
Collapse
Affiliation(s)
- Thomas B Sundberg
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA
| | - Ramnik J Xavier
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stuart L Schreiber
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alykhan F Shamji
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA.
| |
Collapse
|