1
|
Almeida RA, Ferreira CG, Matos VUS, Nogueira JM, Braga MP, Caldi Gomes L, Jorge EC, Soriani FM, Michel U, Ribas VT. AAV-Mediated Expression of miR-17 Enhances Neurite and Axon Regeneration In Vitro. Int J Mol Sci 2024; 25:9057. [PMID: 39201743 PMCID: PMC11355044 DOI: 10.3390/ijms25169057] [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: 07/10/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Neurodegenerative disorders, including traumatic injuries to the central nervous system (CNS) and neurodegenerative diseases, are characterized by early axonal damage, which does not regenerate in the adult mammalian CNS, leading to permanent neurological deficits. One of the primary causes of the loss of regenerative ability is thought to be a developmental decline in neurons' intrinsic capability for axon growth. Different molecules are involved in the developmental loss of the ability for axon regeneration, including many transcription factors. However, the function of microRNAs (miRNAs), which are also modulators of gene expression, in axon re-growth is still unclear. Among the various miRNAs recently identified with roles in the CNS, miR-17, which is highly expressed during early development, emerges as a promising target to promote axon regeneration. Here, we used adeno-associated viral (AAV) vectors to overexpress miR-17 (AAV.miR-17) in primary cortical neurons and evaluate its effects on neurite and axon regeneration in vitro. Although AAV.miR-17 had no significant effect on neurite outgrowth and arborization, it significantly enhances neurite regeneration after scratch lesion and axon regeneration after axotomy of neurons cultured in microfluidic chambers. Target prediction and functional annotation analyses suggest that miR-17 regulates gene expression associated with autophagy and cell metabolism. Our findings suggest that miR-17 promotes regenerative response and thus could mitigate neurodegenerative effects.
Collapse
Affiliation(s)
- Raquel Alves Almeida
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Carolina Gomes Ferreira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Victor Ulysses Souza Matos
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Julia Meireles Nogueira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Marina Pimenta Braga
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31279-901, Brazil (F.M.S.)
| | - Lucas Caldi Gomes
- Clinical Department of Neurology, TUM School of Medicine, Rechts der Isar Hospital, Technical University of Munich, 81675 Munich, Germany;
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Frederico Marianetti Soriani
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31279-901, Brazil (F.M.S.)
| | - Uwe Michel
- Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Vinicius Toledo Ribas
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| |
Collapse
|
2
|
Kuczyńska M, Moskot M, Gabig-Cimińska M. Insights into Autophagic Machinery and Lysosomal Function in Cells Involved in the Psoriatic Immune-Mediated Inflammatory Cascade. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0005. [PMID: 38409665 DOI: 10.2478/aite-2024-0005] [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: 10/06/2023] [Accepted: 12/08/2023] [Indexed: 02/28/2024]
Abstract
Impaired autophagy, due to the dysfunction of lysosomal organelles, contributes to maladaptive responses by pathways central to the immune system. Deciphering the immune-inflammatory ecosystem is essential, but remains a major challenge in terms of understanding the mechanisms responsible for autoimmune diseases. Accumulating evidence implicates a role that is played by a dysfunctional autophagy-lysosomal pathway (ALP) and an immune niche in psoriasis (Ps), one of the most common chronic skin diseases, characterized by the co-existence of autoimmune and autoinflammatory responses. The dysregulated autophagy associated with the defective lysosomal system is only one aspect of Ps pathogenesis. It probably cannot fully explain the pathomechanism involved in Ps, but it is likely important and should be seriously considered in Ps research. This review provides a recent update on discoveries in the field. Also, it sheds light on how the dysregulation of intracellular pathways, coming from modulated autophagy and endolysosomal trafficking, characteristic of key players of the disease, i.e., skin-resident cells, as well as circulating immune cells, may be responsible for immune impairment and the development of Ps.
Collapse
Affiliation(s)
- Martyna Kuczyńska
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | - Marta Moskot
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
3
|
Zhang X, Su J, Lin J, Liu L, Wu J, Yuan W, Zhang Y, Chen Q, Su ZJ, Xu G, Sun M, Zhang Y, Chen X, Zhang W. Fu-Zheng-Tong-Luo formula promotes autophagy and alleviates idiopathic pulmonary fibrosis by controlling the Janus kinase 2/signal transducer and activator of transcription 3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116633. [PMID: 37207878 DOI: 10.1016/j.jep.2023.116633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fu-Zheng-Tong-Luo (FZTL) formula is a Chinese herbal prescription which is used to treat idiopathic pulmonary fibrosis (IPF). We previously reported that the FZTL formula could improve IPF injury in rats; however, the mechanism remains unelucidated. AIM OF THE STUDY To elucidate the effects and mechanisms of the FZTL formula on IPF. MATERIALS AND METHODS The bleomycin-induced pulmonary fibrosis rat model and transforming growth factor-β-induced lung fibroblast model were used. Histological changes and fibrosis formation were detected in the rat model after treatment with the FZTL formula. Furthermore, the effects of the FZTL formula on autophagy and lung fibroblast activation were determined. Moreover, the mechanism of FZTL was explored using transcriptomics analysis. RESULTS We observed that FZTL alleviated IPF injury in rats and inhibited inflammatory responses and fibrosis formation in rats. Moreover, it promoted autophagy and inhibited lung fibroblast activation in vitro. Transcriptomics analysis revealed that FZTL regulates the Janus kinase 2 (JAK)/signal transducer and activator of the transcription 3 (STAT) signaling pathway. The JAK2/STAT3 signaling activator interleukin 6 inhibited the anti-fibroblast activation effect of the FZTL formula. Combined treatment with the JAK2 inhibitor (AZD1480) and autophagy inhibitor (3-methyladenine) did not enhance the antifibrotic effect of FZTL. CONCLUSIONS The FZTL formula can inhibit IPF injury and lung fibroblast activation. Its effects are mediated via the JAK2/STAT3 signaling pathway. The FZTL formula may be a potential complementary therapy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jie Su
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiacheng Lin
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lujiong Liu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiamin Wu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wenli Yuan
- Department of Nephrology Diseases, YueYang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yibao Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qi Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zi Jian Su
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Meng Sun
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yile Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xuan Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wei Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Kuo KL, Chiang CW, Chen YMA, Yu CC, Lee TS. Folic Acid Ameliorates Renal Injury in Experimental Obstructive Nephropathy: Role of Glycine N-Methyltransferase. Int J Mol Sci 2023; 24:ijms24076859. [PMID: 37047834 PMCID: PMC10095475 DOI: 10.3390/ijms24076859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Folic acid exerts both anti-inflammatory and antifibrotic effects. Glycine N-methyltransferase (GNMT), the major folic acid-binding protein in the liver, is a crucial enzyme that regulates the cellular methylation process by maintaining S-adenosylmethionine levels. However, as yet neither the therapeutic effects of folic acid in renal fibrosis nor whether GNMT is involved in these folic acid-associated mechanisms has been investigated. First, the expression of GNMT was examined in human kidneys with or without obstructive nephropathy. Later, wild-type and GNMT knockout (GNMT-/-) mice were subjected to unilateral ureteral obstruction (UUO) and then treated with either folic acid or vehicle for 14 days. Renal tubular injury, inflammation, fibrosis, and autophagy were evaluated by histological analysis and Western blotting. We observed increased expression of GNMT in humans with obstructive nephropathy. Furthermore, UUO significantly increased the expression of GNMT in mice; in addition, it caused renal injury as well as the development of both hydronephrosis and tubular injury. These were all alleviated by folic acid treatment. In contrast, GNMT-/- mice exhibited exacerbated UUO-induced renal injury, but the protective effect of folic acid was not observed in GNMT-/- mice. We propose a novel role for folic acid in the treatment of renal fibrosis, which indicates that GNMT may be a therapeutic target.
Collapse
Affiliation(s)
- Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231405, Taiwan
- School of Medicine, Buddhist Tzu Chi University, Hualien 97004, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Chin-Wei Chiang
- Department of Physiology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Ming Arthur Chen
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei 24205, Taiwan
| | - Chih-Chin Yu
- Division of Urology, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231405, Taiwan
- College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute, Department of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
5
|
Akkoc Y, Dalci K, Karakas HE, Erbil-Bilir S, Yalav O, Sakman G, Celik F, Arikan S, Zeybek U, Ergin M, Akkiz H, Dilege E, Dengjel J, Dogan-Ekici AI, Gozuacik D. Tumor-derived CTF1 (cardiotrophin 1) is a critical mediator of stroma-assisted and autophagy-dependent breast cancer cell migration, invasion and metastasis. Autophagy 2023; 19:306-323. [PMID: 35722965 PMCID: PMC9809961 DOI: 10.1080/15548627.2022.2090693] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved cellular stress response mechanism. Autophagy induction in the tumor microenvironment (stroma) has been shown to support tumor metabolism. However, cancer cell-derived secreted factors that initiate communication with surrounding cells and stimulate autophagy in the tumor microenvironment are not fully documented. We identified CTF1/CT-1 (cardiotrophin 1) as an activator of autophagy in fibroblasts and breast cancer-derived carcinoma-associated fibroblasts (CAFs). We showed that CTF1 stimulated phosphorylation and nuclear translocation of STAT3, initiating transcriptional activation of key autophagy proteins. Additionally, following CTF1 treatment, AMPK and ULK1 activation was observed. We provided evidence that autophagy was important for CTF1-dependent ACTA2/α-SMA accumulation, stress fiber formation and fibroblast activation. Moreover, promotion of breast cancer cell migration and invasion by activated fibroblasts depended on CTF1 and autophagy. Analysis of the expression levels of CTF1 in patient-derived breast cancer samples led us to establish a correlation between CTF1 expression and autophagy in the tumor stroma. In line with our in vitro data on cancer migration and invasion, higher levels of CTF1 expression in breast tumors was significantly associated with lymph node metastasis in patients. Therefore, CTF1 is an important mediator of tumor-stroma interactions, fibroblast activation and cancer metastasis, and autophagy plays a key role in all these cancer-related events.Abbreviations: ACTA2/α-SMA: actin, alpha 2, smooth muscle CAFs: cancer- or carcinoma-associated fibroblasts CNT Ab.: control antibody CNTF: ciliary neurotrophic factor CTF1: cardiotrophin 1 CTF1 Neut. Ab.: CTF1-specific neutralizing antibody GFP-LC3 MEF: GFP-fused to MAP1LC3 protein transgenic MEF LIF: leukemia inhibitory factor IL6: interleukin 6 MEFs: mouse embryonic fibroblasts MEF-WT: wild-type MEFs OSM: oncostatin M TGFB/TGFβ: transforming growth factor beta.
Collapse
Affiliation(s)
- Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey,Department of Biotechnology, Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Kubilay Dalci
- Faculty of Medicine, Department of General Surgery, Çukurova University, Adana, Turkey
| | - Hacer Ezgi Karakas
- Department of Biotechnology, Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Secil Erbil-Bilir
- Department of Biotechnology, Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Orcun Yalav
- Faculty of Medicine, Department of General Surgery, Çukurova University, Adana, Turkey
| | - Gurhan Sakman
- Faculty of Medicine, Department of General Surgery, Çukurova University, Adana, Turkey
| | - Faruk Celik
- Department of Molecular Medicine, Istanbul University Aziz Sancar Institute of Experimental Medicine, Istanbul, Turkey
| | - Soykan Arikan
- Department of General Surgery, Ministry of Health Samatya Training and Research Hospital, Istanbul, Turkey
| | - Umit Zeybek
- Department of Molecular Medicine, Istanbul University Aziz Sancar Institute of Experimental Medicine, Istanbul, Turkey
| | - Melek Ergin
- Faculty of Medicine, Department of Pathology, Çukurova University, Adana, Turkey
| | - Hikmet Akkiz
- Faculty of Medicine, Department of Gastroenterology, Çukurova University, Adana, Turkey
| | - Ece Dilege
- Koç University Hospital, Department of General Surgery, Koç University Medical School, Istanbul, Turkey,School of Medicine, Koç University, Istanbul, Turkey
| | - Joern Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - A. Isin Dogan-Ekici
- School of Medicine, Department of Pathology, Acibadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey,Department of Biotechnology, Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey,School of Medicine, Koç University, Istanbul, Turkey,CONTACT Devrim Gozuacik Koç University School of Medicine, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey; Department of Biotechnology, Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey; School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
6
|
Hu Q, Bian Q, Rong D, Wang L, Song J, Huang HS, Zeng J, Mei J, Wang PY. JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens. Front Bioeng Biotechnol 2023; 11:1110765. [PMID: 36911202 PMCID: PMC9995824 DOI: 10.3389/fbioe.2023.1110765] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Janus kinase/signal transduction and transcription activation (JAK/STAT) pathways were originally thought to be intracellular signaling pathways that mediate cytokine signals in mammals. Existing studies show that the JAK/STAT pathway regulates the downstream signaling of numerous membrane proteins such as such as G-protein-associated receptors, integrins and so on. Mounting evidence shows that the JAK/STAT pathways play an important role in human disease pathology and pharmacological mechanism. The JAK/STAT pathways are related to aspects of all aspects of the immune system function, such as fighting infection, maintaining immune tolerance, strengthening barrier function, and cancer prevention, which are all important factors involved in immune response. In addition, the JAK/STAT pathways play an important role in extracellular mechanistic signaling and might be an important mediator of mechanistic signals that influence disease progression, immune environment. Therefore, it is important to understand the mechanism of the JAK/STAT pathways, which provides ideas for us to design more drugs targeting diseases based on the JAK/STAT pathway. In this review, we discuss the role of the JAK/STAT pathway in mechanistic signaling, disease progression, immune environment, and therapeutic targets.
Collapse
Affiliation(s)
- Qian Hu
- Department of Pharmacy, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Qihui Bian
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Dingchao Rong
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Leiyun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Department of Pharmacy, Wuhan First Hospital, Wuhan, China
| | - Jianan Song
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Hsuan-Shun Huang
- Department of Research, Center for Prevention and Therapy of Gynecological Cancers, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Jun Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Mei
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Peng-Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
GADD34 Ablation Exacerbates Retinal Degeneration in P23H RHO Mice. Int J Mol Sci 2022; 23:ijms232213748. [PMID: 36430227 PMCID: PMC9697375 DOI: 10.3390/ijms232213748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The UPR is sustainably activated in degenerating retinas, leading to translational inhibition via p-eIF2α. Recent findings have demonstrated that ablation of growth arrest and DNA damage-inducible protein 34 (GADD34), a protein phosphatase 1 regulatory subunit permitting translational machinery operation through p-eIF2α elevation, does not impact the rate of translation in fast-degenerating rd16 mice. The current study aimed to validate whether P23H RHO mice degenerating at a slower pace manifest translational attenuation and whether GADD34 ablation impacts the rate of retinal degeneration via further suppression of retinal protein synthesis and apoptotic cell death. For this study, mice were examined with ERG and histological analyses. The molecular assessment was conducted in the naïve and LPS-challenged mice using Western blot and qRT-PCR analyses. Thus, this study demonstrates that the P23H RHO retinas manifest translational attenuation. However, GADD34 ablation resulted in a more prominent p-eIF2a increase without impacting the translation rate. GADD34 deficiency also led to a reduction in scotopic ERG amplitudes and an increased number of TUNEL-positive cells. Molecular analysis revealed that GADD34 deficiency reduces the expression of p-STAT3 and Il-6 while increasing the expression of Tnfa. Overall, the data indicate that GADD34 plays a multifunctional role. Under chronic UPR activation, GADD34 acts as a feedback player, dephosphorylating p-eIF2a, although this role does not seem to be critical. Additionally, GADD34 controls cytokine expression and STAT3 activation. Perhaps these molecular events are particularly important in controlling the pace of retinal degeneration.
Collapse
|
8
|
HEIH Promotes Malignant Progression of Gastric Cancer by Regulating STAT3-Mediated Autophagy and Glycolysis. DISEASE MARKERS 2022; 2022:2634526. [PMID: 36246567 PMCID: PMC9568361 DOI: 10.1155/2022/2634526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022]
Abstract
To study the clinical value of HEIH hyperexpression in gastric cancer and the molecular mechanism of promoting malignant proliferation of gastric cancer cells, qRT-PCR was used to detect the expression of HEIH in gastric cancer and nontumor gastric tissues. HEIH interference sequence was constructed to downregulate HEIH expression in MGC-803 and BGC-823 cell lines. CCK8, clonogenesis, and Transwell assay were used to detect the effects of HEIH on proliferation and invasion of tumor cells. The protein levels of STAT3, p-STAT3, P62, and LC3 were detected by Western blotting. The results showed that HEIH was highly expressed in gastric cancer (P < 0.01). Interference of HEIH expression in MGC-803 and BGC-823 cells reduced the proliferation and invasion of gastric cancer cells, and the results were statistically significant (P < 0.05). HEIH acts as a miRNA sponge for miR-4500. HEIH promotes gastric cancer development by inhibiting miR-4500. STAT3 is a downstream target of miR-4500. HEIH inhibits autophagy and promotes glycolysis. In conclusion, HEIH is highly expressed in gastric cancers. HEIH promotes malignant proliferation and development of gastric cancer cells. HEIH may be a new candidate site for pathological diagnosis and molecular drug therapy for future clinical treatment of gastric cancer.
Collapse
|
9
|
Xu J, Zhang J, Mao QF, Wu J, Wang Y. The Interaction Between Autophagy and JAK/STAT3 Signaling Pathway in Tumors. Front Genet 2022; 13:880359. [PMID: 35559037 PMCID: PMC9086235 DOI: 10.3389/fgene.2022.880359] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/21/2022] [Indexed: 01/30/2023] Open
Abstract
Tumor is one of the important factors affecting human life and health in today’s world, and scientists have studied it extensively and deeply, among which autophagy and JAK/STAT3 signaling pathway are two important research directions. The JAK/STAT3 axis is a classical intracellular signaling pathway that assumes a key role in the regulation of cell proliferation, apoptosis, and vascular neogenesis, and its abnormal cell signaling and regulation are closely related to the occurrence and development of tumors. Therefore, the JAK/STAT3 pathway in tumor cells and various stromal cells in their microenvironment is often considered as an effective target for tumor therapy. Autophagy is a process that degrades cytoplasmic proteins and organelles through the lysosomal pathway. It is a fundamental metabolic mechanism for intracellular degradation. The mechanism of action of autophagy is complex and may play different roles at various stages of tumor development. Altered STAT3 expression has been found to be accompanied by the abnormal autophagy activity in many oncological studies, and the two may play a synergistic or antagonistic role in promoting or inhibiting the occurrence and development of tumors. This article reviews the recent advances in autophagy and its interaction with JAK/STAT3 signaling pathway in the pathogenesis, prevention, diagnosis, and treatment of tumors.
Collapse
Affiliation(s)
- Jiangyan Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinrong Zhang
- Department of Science and Education, Dafeng District People's Hospital, Yancheng, China
| | - Qi-Fen Mao
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuan Wang
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
10
|
Understanding autophagy role in cancer stem cell development. Mol Biol Rep 2022; 49:6741-6751. [PMID: 35277787 DOI: 10.1007/s11033-022-07299-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/30/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of immature cells located in the tumor mass. These cells are responsible for tumor development, proliferation, resistance and spreading. CSCs are characterized by three unique features: the ability to self-renew, differentiation and tumor formation. CSCs are similar to stem cells, but they differ in the malignant phenotype. CSCs become immortal and survive harsh environmental conditions such as hypoxia, starvation and oxidative stress. However, this harsh tumor microenvironment induces the activation of autophagy, which further increases the CSCs stemness profile, and all these features further increase tumorigenicity and metastasis capacity. Autophagy is induced by the extracellular and cellular microenvironment. Hypoxia is one of the most common factors that highly increases the activity of autophagy in CSCs. Therefore, hypoxia-induced autophagy and CSCs proliferation should be elucidated in order to find a novel cure to defeat cancer cells (CSCs and non-CSCs). The remaining challenges to close the gap between the laboratory bench and the development of therapies, to use autophagy against CSCs in patients, could be addressed by adopting a 3D platform to better-mimic the natural environment in which these cells reside. Ultimately allowing to obtain the blueprints for bioprocess scaling up and to develop the production pipeline for safe and cost-effective autophagy-based novel biologics.
Collapse
|
11
|
Yu Y, Ou-Yang WX, Zhang H, Jiang T, Tang L, Tan YF, Luo HY, Xiao ZH, Li SJ. MiR-125b enhances autophagic flux to improve septic cardiomyopathy via targeting STAT3/HMGB1. Exp Cell Res 2021; 409:112842. [PMID: 34563514 DOI: 10.1016/j.yexcr.2021.112842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023]
Abstract
We explore the role of miR-125b in septic cardiomyopathy, focusing on miR-125b/STAT3/HMGB1 axis. CLP mouse model and LPS-stimulated primary rat cardiomyocytes (CMs) and H9C2 cell were used as in vivo and in vitro models of septic cardiomyopathy, respectively. qRT-PCR and western blot were performed to measure expression levels of miR-125b, STAT3, HMGB1, and autophagy-related proteins. MTT assay was employed to examine LPS toxicity. Dual luciferase activity assay and CHIP were performed to validate interactions of miR-125b/STAT3 and STAT3/HMGB1 promoter. Immunostaining was used to assess the level of autophagic flux. ROS level was measured by fluorescence assay. Heart functions were examined via intracoronary Doppler ultrasound. miR-125b was diminished while STAT3 and HMGB1 were elevated in the heart tissue following CLP surgery and in LPS-treated H9C2 cells. LPS treatment up-regulated ROS generation and suppressed autophagic flux. Overexpression of miR-125b mimics or knockdown of STAT3 or HMGB1 alleviated LPS-induced hindrance of autophagic flux and ROS production. miR-125b directly targeted STAT3 mRNA and STAT3 bound with HMGB1 promoter. Overexpression of miR-125b mitigated myocardial dysfunction induced by CLP in vivo. Hyperactivation of STAT3/HMGB1 caused by reduced miR-125b contributes to ROS generation and the hindrance of autophagic flux during septic cardiomyopathy, leading to myocardial dysfunction.
Collapse
Affiliation(s)
- Ying Yu
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Wen-Xian Ou-Yang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hui Zhang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Tao Jiang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Lian Tang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Yan-Fang Tan
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hai-Yan Luo
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Zheng-Hui Xiao
- Emergence Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Shuang-Jie Li
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China.
| |
Collapse
|
12
|
Effect of (-)-Epigallocatechin Gallate on Activation of JAK/STAT Signaling Pathway by Staphylococcal Enterotoxin A. Toxins (Basel) 2021; 13:toxins13090609. [PMID: 34564613 PMCID: PMC8473440 DOI: 10.3390/toxins13090609] [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: 08/01/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcal enterotoxin A (SEA), which is a superantigen toxin protein, binds to cytokine receptor gp130. Gp130 activates intracellular signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. The effects of SEA on the JAK/STAT signaling pathway in mouse spleen cells were examined. After treatment with SEA, mRNA expression levels of interferon gamma (IFN-γ) and suppressor of cytokine-signaling 1 (SOCS1) increased. SEA-induced IFN-γ and SOCS1 expression were decreased by treatment with (-)-epigallocatechin gallate (EGCG). The phosphorylated STAT3, Tyr705, increased significantly in a SEA concentration-dependent manner in mouse spleen cells. Although (-)-3″-Me-EGCG did not inhibit SEA-induced phosphorylated STAT3, EGCG and (-)-4″-Me-EGCG significantly inhibited SEA-induced phosphorylated STAT3. It was thought that the hydroxyl group at position 3 of the galloyl group in the EGCG was responsible for binding to SEA and suppressing SEA-induced phosphorylation of STAT3. Through protein thermal shift assay in vitro, the binding of the gp130 receptor to SEA and the phosphorylation of STAT3 were inhibited by the interaction between EGCG and SEA. As far as we know, this is the first report to document that EGCG inhibits the binding of the gp130 receptor to SEA and the associated phosphorylation of STAT3.
Collapse
|
13
|
Chen L, Wang H, Ge S, Tai S. IL-6/STAT3 pathway is involved in the regulation of autophagy in chronic non-bacterial prostatitis cells, and may be affected by the NLRP3 inflammasome. Ultrastruct Pathol 2021; 45:297-306. [PMID: 34423720 DOI: 10.1080/01913123.2021.1966149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Studies have shown that the cytokine IL-6 plays an important role in the occurrence and development of chronic non-bacterial prostatitis (CNP), but the specific mechanism by which this cytokine regulates CNP is still unclear. At the same time, relevant research have also shown that autophagy is involved in regulating the occurrence and development of inflammation. The possible mechanisms are IL-6/STAT3 signaling pathway and NLRP3 inflammasome. On the basis of establishing the CNP model in rats, we found that IL-6 can regulate autophagy of CNP cells and is associated with the STAT3 pathway and NLRP3 inflammasome. Our results indicate that IL-6 is involved in the regulation of autophagy signaling pathways in CNP. IL-6/STAT3 signaling pathway can suppress cell autophagy pathway in CNP; And the NLRP3 inflammasome may regulate CNP cell autophagy by regulating the IL-6/STAT3 pathway. These findings may provide a new theoretical basis for the pathogenesis of CNP, as well as new ideas and new targets for the treatment and prevention of CNP.
Collapse
Affiliation(s)
- Lidong Chen
- Department Of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Hui Wang
- Department Of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Shengdong Ge
- Department Of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Sheng Tai
- Department Of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
14
|
Qin YR, Ma CQ, Wang DP, Zhang QQ, Liu MR, Zhao HR, Jiang JH, Fang Q. Bilobalide alleviates neuroinflammation and promotes autophagy in Alzheimer's disease by upregulating lincRNA-p21. Am J Transl Res 2021; 13:2021-2040. [PMID: 34017373 PMCID: PMC8129331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
EGb 761 has some protective effects on AD and can improve the cognitive functions of AD mice. However, the underlying molecular mechanisms are unknown. Here, we investigated the function of bilobalide, the effective component of EGb 761, in neuroinflammation and autophagy during AD. LPS-treated BV-2 cells were used as an in vitro model for neuroinflammation. The APP/PS1 AD mouse line was used to examine the function of bilobalide in AD. ELISA and qRT-PCR were used to measure the levels of proinflammatory cytokines, including TNF-α, IL-6 and IL-1β. Western blotting was employed to determine the protein levels of p-p65, iNOS, COX-2, LC3, beclin-1, p62 and p-STAT3. Immunostaining was applied to examine the number of autophagosomes. LPS treatment induced inflammatory responses and inhibited autophagy in BV-2 cells. Bilobalide suppressed LPS-induced neuroinflammation and promoted autophagy. Furthermore, bilobalide treatment increased the lincRNA-p21 levels, which suppressed STAT3 signalling. Knockdown of lincRNA-p21 reversed the effects of bilobalide. Overexpression of lincRNA-p21 promoted autophagy and inhibited neuroinflammation as well while STAT3 inhibitor blocked the effects of si-lincRNA-p21. In vivo experiments revealed that bilobalide improved the learning and memory capabilities of APP/PS1 AD mice. Bilobalide improves the cognitive functions of APP/PS1 AD mice. Mechanistically, bilobalide suppresses inflammatory responses and promotes autophagy possibly by upregulating lincRNA-p21 levels.
Collapse
Affiliation(s)
- Yi-Ren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Chi-Qian Ma
- Department of Cardiology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Da-Peng Wang
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Quan-Quan Zhang
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Mei-Rong Liu
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Hong-Ru Zhao
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Jian-Hua Jiang
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, China
| |
Collapse
|
15
|
Sun Y, Nie W, Qiu B, Guo X, Zhang J, Wei J. Inhibition of microRNA-17 enhances cisplatin-induced apoptosis of human tongue squamous carcinoma cell. J Bioenerg Biomembr 2021; 53:169-176. [PMID: 33462751 DOI: 10.1007/s10863-020-09869-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
MicroRNA-17 (miR-17) was reported to promote cell proliferation and migration of various types of cancers. However, the mechanism remains unclear. This present study was designed to explore the potential mechanism. Downregulation of miR-17 in CAL-27 cells was performed by transfecting anti-miR-27 plasmids. Xenograft tumor model was carried out to detect the effect of inhibition of microRNA-17 on tongue squamous carcinoma growth. MiR-17 inhibition promotes cisplatin-induced apoptosis via regulating the expression of apoptotic molecules. MiR-17 inhibition promotes cisplatin-induced autophagy of CAL-27 cells. Mechanically, miR-17 inhibition promotes apoptosis and autophagy through STAT3 signaling pathway. Xenograft tumor model showed that miR-17 inhibition attenuates tongue squamous carcinoma growth and promotes tongue squamous carcinoma cell apoptosis in vivo. MiR-17 inhibition enhances cisplatin-induced apoptosis of human tongue squamous carcinoma cell. Our study supplies the evidence that miR-17 may serve as the potential target for human tongue squamous carcinoma treatment.
Collapse
Affiliation(s)
- Yang Sun
- Dental Clinic, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China.
| | - Wei Nie
- Dental Department, Cangzhou People's Hospital, No. 7 Qingchi Avence, Cangzhou, 061000, Hebei, China
| | - Bo Qiu
- Dental Clinic, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Xiangjun Guo
- Dental Clinic, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Jian Zhang
- Dental Clinic, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Jianming Wei
- Dental Clinic, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| |
Collapse
|
16
|
Jin KT, Tao XH, Fan YB, Wang SB. Crosstalk between oncolytic viruses and autophagy in cancer therapy. Biomed Pharmacother 2020; 134:110932. [PMID: 33370632 DOI: 10.1016/j.biopha.2020.110932] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses have attracted attention as a promising strategy in cancer therapy owing to their ability to selectively infect and kill tumor cells, without affecting healthy cells. They also exert their anti-tumor effects by releasing immunostimulatory molecules from dying cancer cells. Several regulatory mechanisms, such as autophagy, contribute to the anti-tumor properties of oncolytic viruses. Autophagy is a conserved catabolic process in responses to various stresses, such as nutrient deprivation, hypoxia, and infection that produces energy by lysosomal degradation of intracellular contents. Autophagy can support infectivity and replication of the oncolytic virus and enhance their anti-tumor effects via mediating oncolysis, autophagic cell death, and immunogenic cell death. On the other hand, autophagy can reduce the cytotoxicity of oncolytic viruses by providing survival nutrients for tumor cells. In his review, we summarize various types of oncolytic viruses in clinical trials, their mechanism of action, and autophagy machinery. Furthermore, we precisely discuss the interaction between oncolytic viruses and autophagy in cancer therapy and their combinational effects on tumor cells.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, PR China
| | - Xiao-Hua Tao
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China
| | - Yi-Bin Fan
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China.
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China.
| |
Collapse
|
17
|
Piao JY, Kim SJ, Kim DH, Park JH, Park SA, Han HJ, Na HK, Yoon K, Lee HN, Kim N, Hahm KB, Surh YJ. Helicobacter pylori infection induces STAT3 phosphorylation on Ser727 and autophagy in human gastric epithelial cells and mouse stomach. Sci Rep 2020; 10:15711. [PMID: 32973302 PMCID: PMC7519032 DOI: 10.1038/s41598-020-72594-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/31/2020] [Indexed: 01/05/2023] Open
Abstract
Helicobacter pylori (H. pylori) infection is considered as one of the principal risk factors of gastric cancer. Constitutive activation of the signal transducer and activator of transcription 3 (STAT3) plays an important role in inflammation-associated gastric carcinogenesis. In the canonical STAT3 pathway, phosphorylation of STAT3 on Tyr705 is a major event of STAT3 activation. However, recent studies have demonstrated that STAT3 phosphorylated on Ser727 has an independent function in mitochondria. In the present study, we found that human gastric epithelial AGS cells infected with H. pylori resulted in localization of STAT3 phosphorylated on Ser727 (P-STAT3Ser727), predominantly in the mitochondria. Notably, H. pylori-infected AGS cells exhibited the loss of mitochondrial integrity and increased expression of the microtubule-associated protein light chain 3 (LC3), the autophagosomal membrane-associated protein. Treatment of AGS cells with a mitophagy inducer, carbonyl cyanide 3-chlorophenylhydrazone (CCCP), resulted in accumulation of P-STAT3Ser727 in mitochondria. In addition, the elevated expression and mitochondrial localization of LC3 induced by H. pylori infection were attenuated in AGS cells harboring STAT3 mutation defective in Ser727 phosphorylation (S727A). We also observed that both P-STAT3Ser727 expression and LC3 accumulation were increased in the mitochondria of H. pylori-inoculated mouse stomach.
Collapse
Affiliation(s)
- Juan-Yu Piao
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do, 16227, South Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Sin-Aye Park
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, 31538, South Korea
| | - Hyeong-Jun Han
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, 01133, South Korea
| | - Kichul Yoon
- Department of Internal Medicine and Digestive Disease Research Institute, Wonkwang University Sanbon Hospital, Gunpo, Gyeonggi-do, 15865, South Korea
| | - Ha-Na Lee
- Laboratory of Immunology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Nayoung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Ki Baik Hahm
- Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Gyunggi-do, 13496, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea. .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
18
|
Lv HH, Zhen CX, Liu JY, Shang P. PEITC triggers multiple forms of cell death by GSH-iron-ROS regulation in K7M2 murine osteosarcoma cells. Acta Pharmacol Sin 2020; 41:1119-1132. [PMID: 32132657 PMCID: PMC7468252 DOI: 10.1038/s41401-020-0376-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Phenethyl isothiocyanate (PEITC) is an isothiocyanate that largely exists in cruciferous vegetables and exhibits chemopreventive and chemotherapeutic potential against various cancers. However, it is little known about the molecular mechanisms of its antitumor action against osteosarcoma, which is the second highest cause of cancer-related death in children and adolescents. In this study, we investigated the effects of PEITC on K7M2 murine osteosarcoma both in vitro and in vivo. We found that treatment with PEITC dose-dependently inhibited the viability of K7M2 murine osteosarcoma cells with an IC50 value of 33.49 μM at 24 h. PEITC (1, 15, 30 μM) dose-dependently inhibited the cell proliferation, caused G2/M cell cycle arrest, depleted glutathione (GSH), generated reactive oxygen species (ROS), altered iron metabolism, and triggered multiple forms of cell death, namely ferroptosis, apoptosis, and autophagy in K7M2 cells. We further revealed that PEITC treatment activated MAPK signaling pathway, and ROS generation was a major cause of PEITC-induced cell death. In a syngeneic orthotopic osteosarcoma mouse model, administration of PEITC (30, 60 mg/kg every day, ig, for 24 days) significantly inhibited the tumor growth, but higher dose of PEITC (90 mg/kg every day) compromised its anti-osteosarcoma effect. Histological examination showed that multiple cell death processes were initiated, iron metabolism was altered and MAPK signaling pathway was activated in the tumor tissues. In conclusion, we demonstrate that PEITC induces ferroptosis, autophagy, and apoptosis in K7M2 osteosarcoma cells by activating the ROS-related MAPK signaling pathway. PEITC has promising anti-osteosarcoma activity. This study sheds light on the redox signaling-based chemotherapeutics for cancers.
Collapse
Affiliation(s)
- Huan-Huan Lv
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
- Research Center of Microfluidic Chip for Health Care and Environmental Monitoring, Yangtze River Delta Research Institute of Northwestern Polytechnical University in Taicang, Suzhou, 215400, China
| | - Chen-Xiao Zhen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jun-Yu Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Peng Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China.
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
19
|
Zhang X, Lu T, Ma Y, Li R, Pang Y, Mao H, Liu P. Novel Nanocomplexes Targeting STAT3 Demonstrate Promising Anti-Ovarian Cancer Effects in vivo. Onco Targets Ther 2020; 13:5069-5082. [PMID: 32606729 PMCID: PMC7292488 DOI: 10.2147/ott.s247398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022] Open
Abstract
Background Cationic solid lipid nanoparticles (SLN) have attracted intensive interest as an effective gene delivery system for its high biocompatibility, stability and low cytotoxicity. In our previous study, we successfully prepared SLN-STAT3 decoy ODN complexes and made a primary study on its antitumor behavior in ovarian cancer cells in vitro. However, there is little information available so far about the effect of SLN-STAT3 decoy ODN complexes on ovarian cancer in vivo, either little information about the pharmacological toxicology in vivo. Material and Methods We applied nanotechnology to improve the gene delivery system and synthesize SLN-STAT3 decoy ODN complexes. Xenograft mouse models were established to assess the antitumor effects of SLN-STAT3 decoy ODN on the tumor growth of ovarian cancer in vivo. To analyze the mechanisms of SLN-STAT3 decoy ODN, we investigated apoptosis, autophagy, epithelial–mesenchymal transition (EMT) in tumor tissues of nude mice and investigated the effects and toxicology of SLN-STAT3 decoy ODN complexes on the vital organs of nude mice. Results The results showed that SLN-STAT3 decoy ODN complexes markedly inhibited tumor growth in vivo. SLN-STAT3 decoy ODN complexes could induce cell apoptosis through downregulating Bcl-2, survivin and pro caspase 3, but upregulating Bax and cleaved caspase 3. These complexes could also regulate autophagy through upregulating LC3A-II, LC3B-II and beclin-1, but downregulating p-Akt and p-mTOR. Moreover, these complexes could inhibit cancer cell invasion through reversing EMT. Besides, SLN-STAT3 decoy ODN complexes showed no obvious toxicity on vital organs and hematological parameters of nude mice. Conclusion The molecular mechanisms that SLN-STAT3 decoy ODN complexes inhibit tumor growth involved activating the apoptotic cascade, regulating autophagy, and reversing EMT program; and these complexes showed no obvious toxicity on nude mice. Our study indicated that the nanocomplexes SLN-STAT3 decoy ODN might be a promising therapeutic approach for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Tao Lu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Yanhui Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Rui Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Hongluan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| |
Collapse
|
20
|
El-Gowily AH, Abosheasha MA. Differential mechanisms of autophagy in cancer stem cells: Emphasizing gastrointestinal cancers. Cell Biochem Funct 2020; 39:162-173. [PMID: 32468609 DOI: 10.1002/cbf.3552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/17/2020] [Accepted: 05/03/2020] [Indexed: 12/15/2022]
Abstract
Gastrointestinal (GI) cancers are one of the most common forms of malignancies and still are the most important cause of cancer-related mortality worldwide. Autophagy is a conserved catabolic pathway involving lysosomal degradation and recycling of whole cellular components, which is essential for cellular homeostasis. For instance, it acts as a pivotal intracellular quality control and repair mechanism but also implicated in cell reformation during cell differentiation and development. Indeed, GI cancer stem cells (CSCs) are thought to be responsible for tumour initiation, traditional therapies resistance, metastasis and tumour recurrence. Molecular mechanisms of autophagy in normal vs CSCs gain great interest worldwide. Here, we shed light on the role of autophagy in normal stem cells differentiation for embryonic progression and its role in maintaining the activity and self-renewal capacity of CSCs which offer novel viewpoints on promising cancer therapeutic strategies based on the differential roles of autophagy in CSCs.
Collapse
Affiliation(s)
- Afnan H El-Gowily
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.,Organ and Cell physiology Department, Juntendo University, Tokyo, Japan
| | - Mohammed A Abosheasha
- Cellular Genetics Laboratory, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
21
|
A STAT3 of Addiction: Adipose Tissue, Adipocytokine Signalling and STAT3 as Mediators of Metabolic Remodelling in the Tumour Microenvironment. Cells 2020; 9:cells9041043. [PMID: 32331320 PMCID: PMC7226520 DOI: 10.3390/cells9041043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling of the tumour microenvironment is a major mechanism by which cancer cells survive and resist treatment. The pro-oncogenic inflammatory cascade released by adipose tissue promotes oncogenic transformation, proliferation, angiogenesis, metastasis and evasion of apoptosis. STAT3 has emerged as an important mediator of metabolic remodelling. As a downstream effector of adipocytokines and cytokines, its canonical and non-canonical activities affect mitochondrial functioning and cancer metabolism. In this review, we examine the central role played by the crosstalk between the transcriptional and mitochondrial roles of STAT3 to promote survival and further oncogenesis within the tumour microenvironment with a particular focus on adipose-breast cancer interactions.
Collapse
|
22
|
Billah M, Ridiandries A, Allahwala UK, Mudaliar H, Dona A, Hunyor S, Khachigian LM, Bhindi R. Remote Ischemic Preconditioning induces Cardioprotective Autophagy and Signals through the IL-6-Dependent JAK-STAT Pathway. Int J Mol Sci 2020; 21:ijms21051692. [PMID: 32121587 PMCID: PMC7084188 DOI: 10.3390/ijms21051692] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a cellular process by which mammalian cells degrade and assist in recycling damaged organelles and proteins. This study aimed to ascertain the role of autophagy in remote ischemic preconditioning (RIPC)-induced cardioprotection. Sprague Dawley rats were subjected to RIPC at the hindlimb followed by a 30-min transient blockade of the left coronary artery to simulate ischemia reperfusion (I/R) injury. Hindlimb muscle and the heart were excised 24 h post reperfusion. RIPC prior to I/R upregulated autophagy in the rat heart at 24 h post reperfusion. In vitro, autophagy inhibition or stimulation prior to RIPC, respectively, either ameliorated or stimulated the cardioprotective effect, measured as improved cell viability to mimic the preconditioning effect. Recombinant interleukin-6 (IL-6) treatment prior to I/R increased in vitro autophagy in a dose-dependent manner, activating the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway without affecting the other kinase pathways, such as p38 mitogen-activated protein kinases (MAPK), and glycogen synthase kinase 3 Beta (GSK-3β) pathways. Prior to I/R, in vitro inhibition of the JAK-STAT pathway reduced autophagy upregulation despite recombinant IL-6 pre-treatment. Autophagy is an essential component of RIPC-induced cardioprotection that may upregulate autophagy through an IL-6/JAK-STAT-dependent mechanism, thus identifying a potentially new therapeutic option for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
- School of Life Sciences, Independent University Bangladesh, Dhaka 1229, Bangladesh
- Correspondence:
| | - Anisyah Ridiandries
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| | - Usaid K Allahwala
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| | - Harshini Mudaliar
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Anthony Dona
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Stephen Hunyor
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Ravinay Bhindi
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
23
|
Billah M, Ridiandries A, Rayner BS, Allahwala UK, Dona A, Khachigian LM, Bhindi R. Egr-1 functions as a master switch regulator of remote ischemic preconditioning-induced cardioprotection. Basic Res Cardiol 2019; 115:3. [PMID: 31823016 DOI: 10.1007/s00395-019-0763-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022]
Abstract
Despite improved treatment options myocardial infarction (MI) is still a leading cause of mortality and morbidity worldwide. Remote ischemic preconditioning (RIPC) is a mechanistic process that reduces myocardial infarction size and protects against ischemia reperfusion (I/R) injury. The zinc finger transcription factor early growth response-1 (Egr-1) is integral to the biological response to I/R, as its upregulation mediates the increased expression of inflammatory and prothrombotic processes. We aimed to determine the association and/or role of Egr-1 expression with the molecular mechanisms controlling the cardioprotective effects of RIPC. This study used H9C2 cells in vitro and a rat model of cardiac ischemia reperfusion (I/R) injury. We silenced Egr-1 with DNAzyme (ED5) in vitro and in vivo, before three cycles of RIPC consisting of alternating 5 min hypoxia and normoxia in cells or hind-limb ligation and release in the rat, followed by hypoxic challenge in vitro and I/R injury in vivo. Post-procedure, ED5 administration led to a significant increase in infarct size compared to controls (65.90 ± 2.38% vs. 41.00 ± 2.83%, p < 0.0001) following administration prior to RIPC in vivo, concurrent with decreased plasma IL-6 levels (118.30 ± 4.30 pg/ml vs. 130.50 ± 1.29 pg/ml, p < 0.05), downregulation of the cardioprotective JAK-STAT pathway, and elevated myocardial endothelial dysfunction. In vitro, ED5 administration abrogated IL-6 mRNA expression in H9C2 cells subjected to RIPC (0.95 ± 0.20 vs. 6.08 ± 1.40-fold relative to the control group, p < 0.05), resulting in increase in apoptosis (4.76 ± 0.70% vs. 2.23 ± 0.34%, p < 0.05) and loss of mitochondrial membrane potential (0.57 ± 0.11% vs. 1.0 ± 0.14%-fold relative to control, p < 0.05) in recipient cells receiving preconditioned media from the DNAzyme treated donor cells. This study suggests that Egr-1 functions as a master regulator of remote preconditioning inducing a protective effect against myocardial I/R injury through IL-6-dependent JAK-STAT signaling.
Collapse
Affiliation(s)
- M Billah
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia.
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Life Sciences, Independent University Bangladesh, Dhaka, Bangladesh.
| | - A Ridiandries
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - B S Rayner
- Inflammation Group, Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| | - U K Allahwala
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - A Dona
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - L M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - R Bhindi
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
24
|
Hemorrhagic Shock Sensitized the Diaphragm to Ventilator-Induced Dysfunction through the Activation of IL-6/JAK/STAT Signaling-Mediated Autophagy in Rats. Mediators Inflamm 2019; 2019:3738409. [PMID: 31814800 PMCID: PMC6878811 DOI: 10.1155/2019/3738409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/03/2019] [Indexed: 01/15/2023] Open
Abstract
Mechanical ventilation (MV) is a major life support technique for the management of trauma-associated hemorrhagic shock (HS). Ventilator-induced diaphragm dysfunction (VIDD), one of the most common complications of MV, has been well demonstrated in animal and human studies. However, few data are available concerning the effects of MV on diaphragm function in HS victims. In the present study, we found diaphragm muscle atrophy and weakness in HS but not in healthy animals after 4 hours of MV. The inhibition of autophagy resulted in reduced muscle fiber atrophy and improved forces. In addition, we observed diaphragmatic interleukin- (IL-) 6 overexpression and activation of its downstream signaling JAK/STAT in HS animals after MV, and either the neutralization of IL-6 or the inhibition of the JAK/STAT pathway attenuated autophagy, diaphragm atrophy, and weakness. Importantly, treatment with nonselective antioxidant exerted no protective effects against VIDD in HS animals. In addition, in vitro study showed that exogenous IL-6 was able to induce activation of JAK/STAT signaling and to increase autophagy in C2C12 cells. Moreover, the inhibition of JAK/STAT signaling abolished IL-6-induced cell autophagy. Together, our results suggested that HS sensitized the diaphragm to ventilator-induced atrophy and weakness through the activation of IL-6/JAK/STAT signaling-mediated autophagy in rats.
Collapse
|
25
|
Harhous Z, Booz GW, Ovize M, Bidaux G, Kurdi M. An Update on the Multifaceted Roles of STAT3 in the Heart. Front Cardiovasc Med 2019; 6:150. [PMID: 31709266 PMCID: PMC6823716 DOI: 10.3389/fcvm.2019.00150] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a signaling molecule and transcription factor that plays important protective roles in the heart. The protection mediated by STAT3 is attributed to its genomic actions as a transcription factor and other non-genomic roles targeting mitochondrial function and autophagy. As a transcription factor, STAT3 upregulates genes that are anti-oxidative, anti-apoptotic, and pro-angiogenic, but suppresses anti-inflammatory and anti-fibrotic genes. Its suppressive effects on gene expression are achieved through competing with other transcription factors or cofactors. STAT3 is also linked to the modification of mRNA expression profiles in cardiac cells by inhibiting or inducing miRNA. In addition to these genomic roles, STAT3 is suggested to function protectively in mitochondria, where it regulates ROS production, in part by regulating the activities of the electron transport chain complexes, although our recent evidence calls this role into question. Nonetheless, STAT3 is a key player known to be activated in the cardioprotective ischemic conditioning protocols. Through these varied roles, STAT3 participates in various mechanisms that contribute to cardioprotection against different heart pathologies, including myocardial infarction, hypertrophy, diabetic cardiomyopathy, and peripartum cardiomyopathy. Understanding how STAT3 is involved in the protective mechanisms against these different cardiac pathologies could lead to novel therapeutic strategies to treat them.
Collapse
Affiliation(s)
- Zeina Harhous
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Michel Ovize
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Gabriel Bidaux
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
26
|
Plasticity of High-Density Neutrophils in Multiple Myeloma is Associated with Increased Autophagy Via STAT3. Int J Mol Sci 2019; 20:ijms20143548. [PMID: 32565533 PMCID: PMC6678548 DOI: 10.3390/ijms20143548] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 01/16/2023] Open
Abstract
In both monoclonal gammopathy of uncertain significance (MGUS) and multiple myeloma (MM) patients, immune functions are variably impaired, and there is a high risk of bacterial infections. Neutrophils are the most abundant circulating leukocytes and constitute the first line of host defense. Since little is known about the contribution of autophagy in the neutrophil function of MGUS and MM patients, we investigated the basal autophagy flux in freshly sorted neutrophils of patients and tested the plastic response of healthy neutrophils to soluble factors of MM. In freshly sorted high-density neutrophils obtained from patients with MGUS and MM or healthy subjects, we found a progressive autophagy trigger associated with soluble factors circulating in both peripheral blood and bone marrow, associated with increased IFNγ and pSTAT3S727. In normal high-density neutrophils, the formation of acidic vesicular organelles, a morphological characteristic of autophagy, could be induced after exposure for three hours with myeloma conditioned media or MM sera, an effect associated with increased phosphorylation of STAT3-pS727 and reverted by treatment with pan-JAK2 inhibitor ruxolitinib. Taken together, our data suggest that soluble factors in MM can trigger contemporary JAK2 signaling and autophagy in neutrophils, targetable with ruxolitinib.
Collapse
|
27
|
Merkley SD, Chock CJ, Yang XO, Harris J, Castillo EF. Modulating T Cell Responses via Autophagy: The Intrinsic Influence Controlling the Function of Both Antigen-Presenting Cells and T Cells. Front Immunol 2018; 9:2914. [PMID: 30619278 PMCID: PMC6302218 DOI: 10.3389/fimmu.2018.02914] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a homeostatic and inducible process affecting multiple aspects of the immune system. This intrinsic cellular process is involved in MHC-antigen (Ag) presentation, inflammatory signaling, cytokine regulation, and cellular metabolism. In the context of T cell responses, autophagy has an influential hand in dictating responses to self and non-self by controlling extrinsic factors (e.g., MHC-Ag, cytokine production) in antigen-presenting cells (APC) and intrinsic factors (e.g., cell signaling, survival, cytokine production, and metabolism) in T cells. These attributes make autophagy an attractive therapeutic target to modulate T cell responses. In this review, we examine the impact autophagy has on T cell responses by modulating multiple aspects of APC function; the importance of autophagy in the activation, differentiation and homeostasis of T cells; and discuss how the modulation of autophagy could influence T cell responses.
Collapse
Affiliation(s)
- Seth D Merkley
- Clinical and Translational Science Center, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - Cameron J Chock
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Albuquerque, NM, United States.,Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Albuquerque, NM, United States
| | - James Harris
- Rheumatology Group, Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Clayton, VIC, Australia
| | - Eliseo F Castillo
- Clinical and Translational Science Center, University of New Mexico Health Sciences Albuquerque, NM, United States.,Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Albuquerque, NM, United States.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine Albuquerque, NM, United States
| |
Collapse
|
28
|
Relaño-Ginés A, Lehmann S, Brillaud E, Belondrade M, Casanova D, Hamela C, Vincent C, Poupeau S, Sarniguet J, Alvarez T, Arnaud JD, Maurel JC, Crozet C. Lithium as a disease-modifying agent for prion diseases. Transl Psychiatry 2018; 8:163. [PMID: 30135493 PMCID: PMC6105724 DOI: 10.1038/s41398-018-0209-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 06/12/2018] [Accepted: 06/20/2018] [Indexed: 12/12/2022] Open
Abstract
Prion diseases still remain incurable despite multiple efforts to develop a treatment. Therefore, it is important to find strategies to at least reduce the symptoms. Lithium has been considered as a neuroprotective agent for years, and the objective of this preclinical study was to evaluate the efficacy of lithium delivered as a water-in-oil microemulsion (Aonys®). This delivery system allows using low doses of lithium and to avoid the toxicity observed in chronic treatments. C57BL/6J mice were intracranially inoculated with ME7 prion-infected brain homogenates and then were treated with lithium from day 90 post inoculation until their death. Lithium was administered at traditional doses (16 mg/kg/day) by the gavage route and at lower doses (40 or 160 µg/kg/day; Aonys®) by the rectal mucosa route. Low doses of lithium (Aonys®) improved the survival of prion-inoculated mice, and also decreased vacuolization, astrogliosis, and neuronal loss compared with controls (vehicle alone). The extent of the protective effects in mice treated with low-dose lithium was comparable or even higher than what was observed in mice that received lithium at the traditional dose. These results indicate that lithium administered using this innovative delivery system could represent a potential therapeutic approach not only for prion diseases but also for other neurodegenerative diseases.
Collapse
Affiliation(s)
- A. Relaño-Ginés
- 0000 0001 2097 0141grid.121334.6Institut de Médecine Régénératrice et de Biothérapie (I.M.R.B.), Physiopathologie, diagnostic et thérapie cellulaire des affections neurodégénératives—Institut National de la Santé et de la Recherche Médicale Université de Montpellier U1183 Centre Hospitalo, Universitaire de Montpellier, Montpellier, France ,grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| | - S. Lehmann
- 0000 0001 2097 0141grid.121334.6Institut de Médecine Régénératrice et de Biothérapie (I.M.R.B.), Physiopathologie, diagnostic et thérapie cellulaire des affections neurodégénératives—Institut National de la Santé et de la Recherche Médicale Université de Montpellier U1183 Centre Hospitalo, Universitaire de Montpellier, Montpellier, France ,grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| | - E. Brillaud
- Medesis Pharma SA, Avenue du Golf, Baillargues, France
| | - M. Belondrade
- grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| | - D. Casanova
- grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| | - C. Hamela
- grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| | - C. Vincent
- Medesis Pharma SA, Avenue du Golf, Baillargues, France
| | - S. Poupeau
- Medesis Pharma SA, Avenue du Golf, Baillargues, France
| | - J. Sarniguet
- Medesis Pharma SA, Avenue du Golf, Baillargues, France
| | - T. Alvarez
- 0000 0001 2097 0141grid.121334.6Etablissement Confiné d’Expérimentation BioCampus, Université Montpellier, Campus Triolet, Bâtiment 53, CECEMA, Montpellier, France
| | - J. D. Arnaud
- 0000 0001 2097 0141grid.121334.6Etablissement Confiné d’Expérimentation BioCampus, Université Montpellier, Campus Triolet, Bâtiment 53, CECEMA, Montpellier, France
| | - J. C. Maurel
- Medesis Pharma SA, Avenue du Golf, Baillargues, France
| | - C. Crozet
- 0000 0001 2097 0141grid.121334.6Institut de Médecine Régénératrice et de Biothérapie (I.M.R.B.), Physiopathologie, diagnostic et thérapie cellulaire des affections neurodégénératives—Institut National de la Santé et de la Recherche Médicale Université de Montpellier U1183 Centre Hospitalo, Universitaire de Montpellier, Montpellier, France ,grid.433120.7Institut de Génétique Humaine, Centre National de la Recherche Scientifique-UPR1142, Montpellier, France
| |
Collapse
|
29
|
Game-changing restraint of Ros-damaged phenylalanine, upon tumor metastasis. Cell Death Dis 2018; 9:140. [PMID: 29396431 PMCID: PMC5833805 DOI: 10.1038/s41419-017-0147-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 12/16/2022]
Abstract
An abrupt increase in metastatic growth as a consequence of the removal of primary tumors suggests that the concomitant resistance (CR) phenomenon might occur in human cancer. CR occurs in murine tumors and ROS-damaged phenylalanine, meta-tyrosine (m-Tyr), was proposed as the serum anti-tumor factor primarily responsible for CR. Herein, we demonstrate for the first time that CR happens in different experimental human solid tumors (prostate, lung anaplastic, and nasopharyngeal carcinoma). Moreover, m-Tyr was detected in the serum of mice bearing prostate cancer (PCa) xenografts. Primary tumor growth was inhibited in animals injected with m-Tyr. Further, the CR phenomenon was reversed when secondary implants were injected into mice with phenylalanine (Phe), a protective amino acid highly present in primary tumors. PCa cells exposed to m-Tyr in vitro showed reduced cell viability, downregulated NFκB/STAT3/Notch axis, and induced autophagy; effects reversed by Phe. Strikingly, m-Tyr administration also impaired both, spontaneous metastasis derived from murine mammary carcinomas (4T1, C7HI, and LMM3) and PCa experimental metastases. Altogether, our findings propose m-Tyr delivery as a novel approach to boost the therapeutic efficacy of the current treatment for metastasis preventing the escape from tumor dormancy.
Collapse
|
30
|
Granato M, Gilardini Montani MS, Santarelli R, D'Orazi G, Faggioni A, Cirone M. Apigenin, by activating p53 and inhibiting STAT3, modulates the balance between pro-apoptotic and pro-survival pathways to induce PEL cell death. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:167. [PMID: 29179721 PMCID: PMC5704516 DOI: 10.1186/s13046-017-0632-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/10/2017] [Indexed: 12/19/2022]
Abstract
Background Apigenin is a flavonoid widely distributed in plant kingdom that exerts cytotoxic effects against a variety of solid and haematological cancers. In this study, we investigated the effect of apigenin against primary effusion lymphoma (PEL), a KSHV-associated B cell lymphoma characterized by a very aggressive behavior, displaying constitutive activation of STAT3 as well as of other oncogenic pathways and harboring wtp53. Methods Cell death was assessed by trypan blue exclusion assay, FACS analysis as well as by biochemical studies. The latter were also utilized to detect the occurrence of autophagy and the molecular mechanisms leading to the activation of both processes by apigenin. FACS analysis was used to measure the intracellular ROS utilizing DCFDA. Results We show that apigenin induced PEL cell death and autophagy along with reduction of intracellular ROS. Mechanistically, apigenin activated p53 that induced catalase, a ROS scavenger enzyme, and inhibited STAT3, the most important pro-survival pathway in PEL, as assessed by p53 silencing. On the other hand, STAT3 inhibition by apigenin resulted in p53 activation, since STAT3 negatively influences p53 activity, highlighting a regulatory loop between these two pathways that modulates PEL cell death/survival. Conclusion The findings of this study demonstrate that apigenin may modulate pro-apoptotic and pro-survival pathways representing a valid therapeutic strategy against PEL.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | | | - Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
31
|
Botbol Y, Guerrero-Ros I, Macian F. Key roles of autophagy in regulating T-cell function. Eur J Immunol 2017; 46:1326-34. [PMID: 27151577 DOI: 10.1002/eji.201545955] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/11/2016] [Accepted: 05/02/2016] [Indexed: 01/06/2023]
Abstract
In the past 10 years, autophagy has emerged as a crucial regulator of T-cell homeostasis, activation, and differentiation. Through the ability to adjust the cell's proteome in response to different stimuli, different forms of autophagy have been shown to control T-cell homeostasis and survival. Autophagic processes can also determine the magnitude of the T-cell response to TCR engagement, by regulating the cellular levels of specific signaling intermediates and modulating the metabolic output in activated T cells. In this review we will examine the mechanisms that control autophagy activity in T cells, such as ROS signaling and signaling through common gamma-chain cytokine receptors, and the different aspect of T-cell biology, including T-cell survival, effector cell function, and generation of memory, which can be regulated by autophagy.
Collapse
Affiliation(s)
- Yair Botbol
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
32
|
Adiponectin deficiency rescues high-fat diet-induced hepatic injury, apoptosis and autophagy loss despite persistent steatosis. Int J Obes (Lond) 2017; 41:1403-1412. [PMID: 28559541 DOI: 10.1038/ijo.2017.128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/01/2017] [Accepted: 05/16/2017] [Indexed: 02/06/2023]
Abstract
Background &aims:Low levels of adiponectin (APN), an adipose-derived adipokine, are associated with obesity and non-alcoholic steatohepatitis although its role in high-fat diet-induced hepatic injury and steatosis remains unclear. Here we hypothesized that APN deficiency alters fat diet-induced hepatic function. To this end, we examined the effect of APN deficiency on high-fat diet-induced hepatic injury, apoptosis and steatosis. METHODS Adult wild type and APN knockout mice were fed a low- or high-fat diet for 20 weeks. Serum levels of liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), cholesterol, hepatic triglycerides, steatosis, pro-inflammatory cytokines, apoptosis and autophagy were examined. RESULTS High-fat feeding led to elevated body (48.2%) and liver weights (18.8%), increased levels of ALT (87.8%), serum cholesterol (104.4%), hepatic triglycerides (305.6%) and hepatic fat deposition as evidenced by Oil Red O staining, along with a reduced AST/ALT ratio and unchanged AST. Although APN knockout itself did not affect hepatic function and morphology, it reconciled fat diet-induced hepatic injury (P<0.05 vs WT-HF group) without reversing changes in body and liver weights, serum cholesterol and hepatic steatosis. In addition, fat diet intake promoted AMPK phosphorylation, p62 accumulation and apoptosis, including elevated Bax and cleaved Caspase-3 and downregulated Bcl-2, along with suppressed phosphorylation of Akt, STAT3 and JNK, and the autophagy makers Atg7, Beclin-1 and LC3B (P<0.05 vs WT-LF group) without affecting hepatic interlelukin-6 and tumor necrosis factor-α levels, the effects were reversed or significantly attenuated by APN knockout (P<0.05 vs WT-HF group). In vitro study using HepG2 cells revealed that STAT3 activation rescued palmitic acid-induced cell injury whereas STAT3 inhibition nullified APN knockdown-offered beneficial effects. CONCLUSIONS Our results revealed that high-fat diet intake promotes hepatic steatosis, apoptosis and interrupted autophagy. APN knockout elicits protective effect against hepatic injury possibly associated with autophagy regulation despite persistent hepatic steatosis.
Collapse
|
33
|
Yang Y, Hu W, Di S, Ma Z, Fan C, Wang D, Jiang S, Li Y, Zhou Q, Li T, Luo E. Tackling myocardial ischemic injury: the signal transducer and activator of transcription 3 (STAT3) at a good site. Expert Opin Ther Targets 2017; 21:215-228. [PMID: 28001439 DOI: 10.1080/14728222.2017.1275566] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Myocardial ischemia is one of the main causes of cardiac remodeling and heart failure. As a highly evolutionarily conserved pathway, the signal transducer and activator of transcription 3 (STAT3) signaling controls intercellular communication, signaling transduction and gene transcription. Interestingly, STAT3 signaling has been demonstrated to take part in myocardial ischemia. Furthermore, activation of STAT3 signaling contributes to the protective efficacy of various interventions, including pharmacological and non-pharmacological treatment of myocardial ischemic injury. Areas covered: We first introduce the protective mechanisms of STAT3. We then discuss STAT3 signaling in various cells and the roles of STAT3 in myocardial processes during myocardial ischemia. Finally, the roles of STAT3 in myocardial ischemia and the upstream regulators of STAT3 activation are summarized. Expert opinion: In various animal experiments, STAT3 has been demonstrated to take part in myocardial responses to myocardial ischemic injury and to be activated during various modes of protection against myocardial ischemia and ischemia/reperfusion (I/R) injury. However, further clinical evidence on the role of STAT3 in such protection is needed. Treatments targeting STAT3 as a means of reducing myocardial ischemic injury need to be tested in a clinical setting. Furthermore, biotechnology can be used to develop effective drugs for this purpose.
Collapse
Affiliation(s)
- Yang Yang
- a Department of Biomedical Engineering , The Fourth Military Medical University , Xi'an , China
- b Department of Thoracic and Cardiovascular Surgery , Affiliated Drum Tower Hospital of Nanjing University Medical School , Nanjing , Jiangsu , China
| | - Wei Hu
- a Department of Biomedical Engineering , The Fourth Military Medical University , Xi'an , China
| | - Shouyin Di
- c Department of Thoracic Surgery , Tangdu Hospital, The Fourth Military Medical University , Xi'an , China
| | - Zhiqiang Ma
- c Department of Thoracic Surgery , Tangdu Hospital, The Fourth Military Medical University , Xi'an , China
| | - Chongxi Fan
- c Department of Thoracic Surgery , Tangdu Hospital, The Fourth Military Medical University , Xi'an , China
| | - Dongjin Wang
- b Department of Thoracic and Cardiovascular Surgery , Affiliated Drum Tower Hospital of Nanjing University Medical School , Nanjing , Jiangsu , China
| | - Shuai Jiang
- d Department of Aerospace Medicine , The Fourth Military Medical University , Xi'an , China
| | - Yue Li
- a Department of Biomedical Engineering , The Fourth Military Medical University , Xi'an , China
| | - Qing Zhou
- b Department of Thoracic and Cardiovascular Surgery , Affiliated Drum Tower Hospital of Nanjing University Medical School , Nanjing , Jiangsu , China
| | - Tian Li
- a Department of Biomedical Engineering , The Fourth Military Medical University , Xi'an , China
| | - Erping Luo
- a Department of Biomedical Engineering , The Fourth Military Medical University , Xi'an , China
| |
Collapse
|
34
|
Ma Y, Zhang X, Xu X, Shen L, Yao Y, Yang Z, Liu P. STAT3 Decoy Oligodeoxynucleotides-Loaded Solid Lipid Nanoparticles Induce Cell Death and Inhibit Invasion in Ovarian Cancer Cells. PLoS One 2015; 10:e0124924. [PMID: 25923701 PMCID: PMC4414561 DOI: 10.1371/journal.pone.0124924] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/10/2015] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the synthesis of multi-functional nanoparticles have opened up tremendous opportunities for the targeted delivery of genes of interest. Cationic solid lipid nanoparticles (SLN) can efficiently bind nucleic acid molecules and transfect genes in vitro. Few reports have combined SLN with therapy using decoy oligodeoxynucleotides (ODN). In the present study, we prepared SLN to encapsulate STAT3 decoy ODN; then, the properties and in vitro behavior of SLN-STAT3 decoy ODN complexes were investigated. SLN-STAT3 decoy ODN complexes were efficiently taken up by human ovarian cancer cells and significantly suppressed cell growth. Blockage of the STAT3 pathway by SLN-STAT3 decoy ODN complexes resulted in an evident induction of cell death, including apoptotic and autophagic death. The mechanism involved the increased expression of cleaved caspase 3, Bax, Beclin-1 and LC3-II and reduced expression of Bcl-2, pro-caspase 3, Survivin, p-Akt and p-mTOR. In addition, SLN-STAT3 decoy ODN complexes inhibited cell invasion by up-regulating E-cadherin expression and down-regulating Snail and MMP-9 expression. These findings confirmed that SLN as STAT3 decoy ODN carriers can induce cell death and inhibit invasion of ovarian cancer cells. We propose that SLN represent a potential approach for targeted gene delivery in cancer therapy.
Collapse
Affiliation(s)
- Yanhui Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaolei Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoxuan Xu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Liang Shen
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated with Shandong University, Jinan, Shandong, China
| | - Yao Yao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Ziyan Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
35
|
Qi Y, Zhang M, Li H, Frank JA, Dai L, Liu H, Zhang Z, Wang C, Chen G. Autophagy inhibition by sustained overproduction of IL6 contributes to arsenic carcinogenesis. Cancer Res 2014; 74:3740-52. [PMID: 24830721 DOI: 10.1158/0008-5472.can-13-3182] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chronic inflammation has been implicated as an etiologic factor in cancer, whereas autophagy may help preserve cancer cell survival but exert anti-inflammatory effects. How these phenomenas interact during carcinogenesis remains unclear. We explored this question in a human bronchial epithelial cell-based model of lung carcinogenesis that is mediated by subchronic exposure to arsenic. We found that sustained overexpression of the pro-inflammatory IL6 promoted arsenic-induced cell transformation by inhibiting autophagy. Conversely, strategies to enhance autophagy counteracted the effect of IL6 in the model. These findings were confirmed and extended in a mouse model of arsenic-induced lung cancer. Mechanistic investigations suggested that mTOR inhibition contributed to the activation of autophagy, whereas IL6 overexpression was sufficient to block autophagy by supporting Beclin-1/Mcl-1 interaction. Overall, our findings argued that chronic inflammatory states driven by IL6 could antagonize autophagic states that may help preserve cancer cell survival and promote malignant progression, suggesting a need to uncouple inflammation and autophagy controls to enable tumor progression.
Collapse
Affiliation(s)
- Yuanlin Qi
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology, Biochemistry and Molecular Biology and
| | - Mingfang Zhang
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology, Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Hui Li
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology
| | - Jacqueline A Frank
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology
| | - Lu Dai
- Graduate Center for Toxicology, University of Kentucky College of Medicine
| | - Huijuan Liu
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology
| | - Zhuo Zhang
- Graduate Center for Toxicology, University of Kentucky College of Medicine
| | - Chi Wang
- Biostatistics Shared Resource Facility, Markey Cancer Center and Department of Biostatistics, University of Kentucky College of Public Health, Lexington, Kentucky; and Departments of
| | - Gang Chen
- Authors' Affiliations: Department of Molecular & Biomedical Pharmacology,
| |
Collapse
|