1
|
Hu Z, Wang J, Pan T, Li X, Tao C, Wu Y, Wang X, Zhang Z, Liu Y, Zhang W, Xu C, Wu X, Gu Q, Fan Y, Qian H, Mugisha A, Yuan S, Liu Q, Xie P. The Exosome-Transmitted lncRNA LOC100132249 Induces Endothelial Dysfunction in Diabetic Retinopathy. Diabetes 2023; 72:1307-1319. [PMID: 37347724 DOI: 10.2337/db22-0435] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
Diabetic retinopathy (DR), one of the most common microangiopathic complications in diabetes, causes severe visual damage among working-age populations. Retinal vascular endothelial cells, the key cell type in DR pathogenesis, are responsible for abnormal retinal angiogenesis in advanced stages of DR. The roles of exosomes in DR have been largely unknown. In this study, we report the first evidence that exosomes derived from the vitreous humor of patients with proliferative DR (PDR-exo) promote proliferation, migration, and tube formation of human retinal vascular endothelial cells (HRVECs). We identified long noncoding RNA (lncRNA) LOC100132249 enrichment in PDR-exo via high-throughput sequencing. This lncRNA, also mainly derived from HRVECs, promoted angiogenesis both in vitro and in vivo. Mechanistically, LOC100132249 acted as a competing endogenous sponge of miRNA-199a-5p (miR-199a-5p), thus regulating the endothelial-mesenchymal transition promoter SNAI1 via activation of the Wnt/β-catenin pathway and ultimately resulting in endothelial dysfunction. In conclusion, our findings underscored the pathogenic role of endothelial-derived exosomes via the LOC100132249/miR-199a-5p/SNAI1 axis in DR angiogenesis and may shed light on new therapeutic strategies for future treatment of DR. ARTICLE HIGHLIGHTS This study provides the first evidence that exosomes derived from vitreous humor from patients with proliferative diabetic retinopathy participate in angiogenesis. The findings demonstrate an unreported long noncoding RNA (lncRNA), LOC100132249, by exosomal sequencing of vitreous humor. The newly found lncRNA LOC100132249, mainly derived from endothelial cells, promotes angiogenesis via an miRNA-199a-5p/SNAI1/Wnt/β-catenin axis in a pro-endothelial-mesenchymal transition manner.
Collapse
Affiliation(s)
- Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingfan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Pan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xinsheng Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Tao
- MOE Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, China
| | - Yan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xingxing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengyu Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Weiwei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Changlin Xu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinjing Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinyuan Gu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Fan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huiming Qian
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aime Mugisha
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Yastrebova MA, Khamidullina AI, Tatarskiy VV, Scherbakov AM. Snail-Family Proteins: Role in Carcinogenesis and Prospects for Antitumor Therapy. Acta Naturae 2021; 13:76-90. [PMID: 33959388 PMCID: PMC8084295 DOI: 10.32607/actanaturae.11062] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
The review analyzes Snail family proteins, which are transcription factors involved in the regulation of the epithelial-mesenchymal transition (EMT) of tumor cells. We describe the structure of these proteins, their post-translational modification, and the mechanisms of Snail-dependent regulation of genes. The role of Snail proteins in carcinogenesis, invasion, and metastasis is analyzed. Furthermore, we focus on EMT signaling mechanisms involving Snail proteins. Next, we dissect Snail signaling in hypoxia, a condition that complicates anticancer treatment. Finally, we offer classes of chemical compounds capable of down-regulating the transcriptional activity of Snails. Given the important role of Snail proteins in cancer biology and the potential for pharmacological inhibition, Snail family proteins may be considered promising as therapeutic targets.
Collapse
Affiliation(s)
- M. A. Yastrebova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - A. I. Khamidullina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - V. V. Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| | - A. M. Scherbakov
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| |
Collapse
|
3
|
Discovery of derivatives of 6(7)-amino-3-phenylquinoxaline-2-carbonitrile 1,4-dioxides: novel, hypoxia-selective HIF-1α inhibitors with strong antiestrogenic potency. Bioorg Chem 2020; 104:104324. [DOI: 10.1016/j.bioorg.2020.104324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022]
|
4
|
Volkova YA, Kozlov AS, Kolokolova MK, Uvarov DY, Gorbatov SA, Andreeva OE, Scherbakov AM, Zavarzin IV. Steroidal N-Sulfonylimidates: Synthesis and biological evaluation in breast cancer cells. Eur J Med Chem 2019; 179:694-706. [DOI: 10.1016/j.ejmech.2019.06.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023]
|
5
|
Novel steroidal 1,3,4-thiadiazines: Synthesis and biological evaluation in androgen receptor-positive prostate cancer 22Rv1 cells. Bioorg Chem 2019; 91:103142. [PMID: 31400555 DOI: 10.1016/j.bioorg.2019.103142] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022]
Abstract
A flexible approach to previously unknown spirofused and linked 1,3,4-thiadiazine derivatives of steroids with selective control of heterocyclization patterns is disclosed. (N-Arylcarbamoyl)spiroandrostene-17,6' [1,3,4]thiadiazines and (N-arylcarbamoyl)17-[1',3',4']thiadiazine-substituted androstenes, novel types of heterosteroids, were prepared from 16β,17β-epoxypregnenolone and 21-bromopregna-5,16-dien-20-one in good to high yields by the treatment with oxamic acid thiohydrazides. The synthesized compounds were screened for antiproliferative activity against the human androgen receptor-positive prostate cancer cell line 22Rv1. Most of (N-arylcarbamoyl)17-[1',3',4']thiadiazine-substituted androstenes exhibit better antiproliferative potency (IC50 = 2.1-6.6 µM) than the antiandrogen bicalutamide. Compounds 7d with IC50 = 3.0 μM and 7j with IC50 = 2.1 μM proved to be the most active in the series under study. Lead synthesized compound 7j downregulates AR expression and activity in 22Rv1 cells. NF-κB activity is also blocked in 7j-treated 22Rv1 cells. Apoptosis is considered as a possible mechanism of 7j-induced cell death.
Collapse
|
6
|
Al-Qassab Y, Grassilli S, Brugnoli F, Vezzali F, Capitani S, Bertagnolo V. Protective role of all-trans retinoic acid (ATRA) against hypoxia-induced malignant potential of non-invasive breast tumor derived cells. BMC Cancer 2018; 18:1194. [PMID: 30497437 PMCID: PMC6267073 DOI: 10.1186/s12885-018-5038-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background The presence of hypoxic areas is common in all breast lesions but no data clearly correlate low oxygenation with the acquisition of malignant features by non-invasive cells, particularly by cells from ductal carcinoma in situ (DCIS), the most frequently diagnosed tumor in women. Methods By using a DCIS-derived cell line, we evaluated the effects of low oxygen availability on malignant features of non-invasive breast tumor cells and the possible role of all-trans retinoic acid (ATRA), a well-known anti-leukemic drug, in counteracting the effects of hypoxia. The involvement of the β2 isoform of PI-PLC (PLC-β2), an ATRA target in myeloid leukemia cells, was also investigated by specific modulation of the protein expression. Results We demonstrated that moderate hypoxia is sufficient to induce, in DCIS-derived cells, motility, epithelial-to-mesenchymal transition (EMT) and expression of the stem cell marker CD133, indicative of their increased malignant potential. Administration of ATRA supports the epithelial-like phenotype of DCIS-derived cells cultured under hypoxia and keeps down the number of CD133 positive cells, abrogating almost completely the effects of poor oxygenation. We also found that the mechanisms triggered by ATRA in non-invasive breast tumor cells cultured under hypoxia is in part mediated by PLC-β2, responsible to counteract the effects of low oxygen availability on CD133 levels. Conclusions Overall, we assigned to hypoxia a role in increasing the malignant potential of DCIS-derived cells and we identified in ATRA, currently used in treatment of acute promyelocytic leukemia (APL), an agonist potentially useful in preventing malignant progression of non-invasive breast lesions showing hypoxic areas.
Collapse
Affiliation(s)
- Yasamin Al-Qassab
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.,College of Medicine, Department of Anatomy, University of Baghdad, Baghdad, Iraq
| | - Silvia Grassilli
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Federica Brugnoli
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Federica Vezzali
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Silvano Capitani
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.,LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Valeria Bertagnolo
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.
| |
Collapse
|
7
|
Scherbakov AM, Zavarzin IV, Vorontsova SK, Hajra A, Andreeva OE, Yadykov AV, Levina IS, Volkova YA, Shirinian VZ. Synthesis and evaluation of the antiproliferative activity of benzylidenes of 16-dehydroprogesterone series. Steroids 2018; 138:91-101. [PMID: 29997047 DOI: 10.1016/j.steroids.2018.06.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/11/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022]
Abstract
Novel benzylidenes (chalcones) of the 16-dehydroprogesterone series have been characterized and their antitumor activity against two breast cancer cell lines was evaluated. Benzylidenes exhibit significant antiproliferative effect on cells and inhibit cell growth in hormone-dependent MCF-7 and hormone-independent MDA-MB-231 breast cancer cell lines. Compound 3d exhibits the highest activity against two breast cancer cell lines, with the IC50 value of about 2 µM. Compounds 3e,m,n display considerable selectivity for hormone-dependent breast cancer cells, with the IC50 value lower than 6 µM. Moreover, these steroidal benzylidenes regulate ERα signaling and reveal p53-independent mechanism of pro-apoptotic action in MCF-7 cells. The new class of antitumor compounds holds promise as the basis for the design of agents for cancer therapy.
Collapse
Affiliation(s)
- Alexander M Scherbakov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, 115478 Moscow, Russia.
| | - Igor V Zavarzin
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| | - Svetlana K Vorontsova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| | - Alakananda Hajra
- Department of Chemistry, Visva-Bharati (A Central University), Santiniketan, India.
| | - Olga E Andreeva
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, 115478 Moscow, Russia.
| | - Anton V Yadykov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| | - Inna S Levina
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| | - Yulia A Volkova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| | - Valerii Z Shirinian
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, 119991 Moscow, Russia.
| |
Collapse
|
8
|
Scherbakov AM, Borunov AM, Buravchenko GI, Andreeva OE, Kudryavtsev IA, Dezhenkova LG, Shchekotikhin AE. Novel Quinoxaline-2-Carbonitrile-1,4-Dioxide Derivatives Suppress HIF1α Activity and Circumvent MDR in Cancer Cells. Cancer Invest 2018; 36:199-209. [PMID: 29624460 DOI: 10.1080/07357907.2018.1453072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A series of 3-aryl/hetarylquinoxaline-2-carbonitrile-1,4-dioxides was synthesized and evaluated against breast cancer cell lines in normoxia and hypoxia. Selected compounds in this series demonstrated better cytotoxicity and comparable hypoxia selectivity than tirapazamine. In contrast to Dox, quinoxaline-1,4-dioxides showed potent cytotoxicity against different MDR cells. Compound 2g inhibits of cancer cell growth through p53-independent mechanisms. Our results showed that compound 2g sensitized MCF-7 cells to metformin in hypoxia. Treatment with 2g results in the increase of ROS accumulation in cancer cells. Compound 2g can be considered as the lead compound for further anticancer drug design, evaluation, and development of new potent antitumor agents.
Collapse
Affiliation(s)
- Alexander M Scherbakov
- a Department of Experimental Tumor Biology , Blokhin N.N. National Medical Research Center of Oncology , Moscow , Russia
| | - Alexander M Borunov
- b Laboratory of Chemical Transformations of Antibiotics , Gause Institute of New Antibiotics , Moscow , Russia.,c Organic Chemistry Department , Mendeleyev University of Chemical Technology of Russia , Moscow , Russia
| | - Galina I Buravchenko
- b Laboratory of Chemical Transformations of Antibiotics , Gause Institute of New Antibiotics , Moscow , Russia.,c Organic Chemistry Department , Mendeleyev University of Chemical Technology of Russia , Moscow , Russia
| | - Olga E Andreeva
- a Department of Experimental Tumor Biology , Blokhin N.N. National Medical Research Center of Oncology , Moscow , Russia
| | - Igor A Kudryavtsev
- a Department of Experimental Tumor Biology , Blokhin N.N. National Medical Research Center of Oncology , Moscow , Russia
| | - Lyubov G Dezhenkova
- b Laboratory of Chemical Transformations of Antibiotics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Andrey E Shchekotikhin
- b Laboratory of Chemical Transformations of Antibiotics , Gause Institute of New Antibiotics , Moscow , Russia.,c Organic Chemistry Department , Mendeleyev University of Chemical Technology of Russia , Moscow , Russia
| |
Collapse
|
9
|
Semina SE, Scherbakov AM, Kovalev SV, Shevchenko VE, Krasil'nikov MA. Horizontal Transfer of Tamoxifen Resistance in MCF-7 Cell Derivates: Proteome Study. Cancer Invest 2017; 35:506-518. [DOI: 10.1080/07357907.2017.1368081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- S. E. Semina
- Laboratory of Molecular Endocrinology, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - A. M. Scherbakov
- Laboratory of Oncoproteomics, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - S. V. Kovalev
- Laboratory of Oncoproteomics, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - V. E. Shevchenko
- Laboratory of Oncoproteomics, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - M. A. Krasil'nikov
- Laboratory of Molecular Endocrinology, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| |
Collapse
|
10
|
Yang C, Zhou Q, Li M, Tong X, Sun J, Qing Y, Sun L, Yang X, Hu X, Jiang J, Yan X, He L, Wan C. Upregulation of CYP2S1 by oxaliplatin is associated with p53 status in colorectal cancer cell lines. Sci Rep 2016; 6:33078. [PMID: 27609465 PMCID: PMC5016804 DOI: 10.1038/srep33078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
Oxaliplatin displays a wide spectrum of antitumor activities and is widely used in the treatment of metastatic colorectal cancer (CRC). However, tumor responses to this agent are variable, and the underlying mechanisms are poorly understood. In the present study, oxaliplatin was found to strongly inhibit the growth of HCT116 cells harboring wild-type p53 but to only weakly inhibit SW480 cells, HT29 cells or p53-/- HCT116 cells, which all lack p53 expression. Administration of oxaliplatin significantly induced p53 accumulation and enhanced expression of CYP2S1 in HCT116 cells with wild-type p53. CYP2S1 knockdown conferred a cell survival advantage after oxaliplatin treatment to cells harboring wild-type p53 in vitro and in vivo. Interestingly, enzyme immunoassays, TOPFlash/FOPFlash reporter activity assays and western blotting analysis demonstrated oxaliplatin-mediated downregulation of PGE2 and Wnt/β-catenin signaling in a manner dependent on p53. Moreover, oxaliplatin treatment of mice with subcutaneous tumor xenografts drastically reduced the volume of wild-type p53 HCT116 tumors but had no effect on isogenic p53-/- HCT116 tumors. These results suggest that oxaliplatin exerts its inhibitory effects in human CRC cells via upregulation of CYP2S1 expression in a p53-dependent manner.
Collapse
Affiliation(s)
- Chao Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- College of Life Science, Anhui Normal University, Anhui Wuhu 241000, China
| | - Qian Zhou
- College of Life Science, Anhui Normal University, Anhui Wuhu 241000, China
| | - Minle Li
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiayi Sun
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yin Qing
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liya Sun
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xuhan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jie Jiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaomei Yan
- School of Life Sciences & Biotechnology, Shanghai JiaoTong University, Shanghai 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
11
|
Scherbakov AM, Gershtein ES, Korotkova EA, Ovchinnikova LK, Ovsii OG, Ermilova VD, Gens GP, Kushlinskii NE. Regulatory Proteins of Epithelial-Mesenchymal Transition and Some Components of VEGF Signaling Pathway in Breast Cancer. Bull Exp Biol Med 2016; 160:802-6. [PMID: 27165081 DOI: 10.1007/s10517-016-3314-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 01/01/2023]
Abstract
Immunohistochemical method was used to assay for Snail family regulatory proteins of epithelial-mesenchymal transition, their NF-κB coactivator, and the components of VEGF signaling pathway (VEGF and its receptors VEGFR1 and VEGFR2) in 157 specimens of breast tumors. Most tumors did not express SNAI1, while 65% tumors demonstrated mid- or high-level SNAI2 expression. There were significant correlations between the expression of SNAI1, SNAI2, and their NF-κB co-activator. Correlation was also detected between expression of Snail and VEGFR1 protein families in the tumors. In addition, the study revealed tumoral co-expression of SNAI2 and VEGFR2. The data attest to coordinated activation of regulatory proteins of epithelial-mesenchymal transition and the major components of VEGF signaling pathway in breast tumors.
Collapse
Affiliation(s)
- A M Scherbakov
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - E S Gershtein
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E A Korotkova
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - L K Ovchinnikova
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - O G Ovsii
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V D Ermilova
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - G P Gens
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N E Kushlinskii
- N. N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
12
|
Qi J, Li T, Bian H, Li F, Ju Y, Gao S, Su J, Ren W, Qin C. SNAI1 promotes the development of HCC through the enhancement of proliferation and inhibition of apoptosis. FEBS Open Bio 2016; 6:326-37. [PMID: 27239445 PMCID: PMC4821356 DOI: 10.1002/2211-5463.12043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/08/2023] Open
Abstract
SNAI1, a zinc‐finger transcription factor, plays an important role in the induction of epithelial–mesenchymal transition (EMT) in various cancers. However, the possible functions of SNAI1 in the proliferation and apoptosis of hepatocellular carcinoma have not been clearly identified. In this study, we investigated the effects and mechanisms of SNAI1 in the proliferation and apoptosis of hepatocellular carcinoma using clinical samples and cell lines. We found that SNAI1 is highly expressed in the tissues of liver cancer compared with adjacent nontumor tissues. SNAI1 is also highly expressed in the hepatoma cell lines HepG2, SMMC‐7721, and BEL‐7402 compared with the human normal liver cell line L02. We also observed that SNAI1 expression was correlated with distal metastasis, incomplete tumor capsule formation, and histological differentiation in hepatocellular carcinoma (HCC). Moreover, we demonstrated that knockdown of SNAI1 via lentiviral vectors of RNAi against SNAI inhibited cell proliferation by inducing G1 arrest, which was accompanied by the downregulation of cyclin D1 but not that of cyclin A. In addition, knockdown of SNAI1 promoted apoptosis by decreasing the expression of Bcl‐2. In conclusion, our findings revealed that SNAI1 is involved in the development of hepatocellular carcinoma via regulating the growth and apoptosis of tumor cells.
Collapse
Affiliation(s)
- Jianni Qi
- Central Laboratory Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Tao Li
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Hongjun Bian
- Department of Emergency Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Feifei Li
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Ying Ju
- Department of Clinical Laboratory Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Shanshan Gao
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Jingran Su
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Wanhua Ren
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Chengyong Qin
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| |
Collapse
|
13
|
Scherbakov AM, Sorokin DV, Tatarskiy VV, Prokhorov NS, Semina SE, Berstein LM, Krasil'nikov MA. The phenomenon of acquired resistance to metformin in breast cancer cells: The interaction of growth pathways and estrogen receptor signaling. IUBMB Life 2016; 68:281-92. [PMID: 26892736 DOI: 10.1002/iub.1481] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 01/10/2016] [Indexed: 01/02/2023]
Abstract
Metformin, a biguanide antidiabetic drug, is used to decrease hyperglycemia in patients with type 2 diabetes. Recently, the epidemiological studies revealed the potential of metformin as an anti-tumor drug for several types of cancer, including breast cancer. Anti-tumor metformin action was found to be mediated, at least in part, via activation of adenosine monophosphate-activated protein kinase (AMPK)-intracellular energy sensor, which inhibits the mammalian target of rapamycin (mTOR) and some other signaling pathways. Nevertheless, some patients can be non-sensitive or resistant to metformin action. Here we analyzed the mechanism of the formation of metformin-resistant phenotype in breast cancer cells and its role in estrogen receptor (ER) regulation. The experiments were performed on the ER-positive MCF-7 breast cancer cells and metformin-resistant MCF-7 subline (MCF-7/M) developed due to long-term metformin treatment. The transcriptional activity of NF-κB and ER was measured by the luciferase reporter gene analysis. The protein expression was determined by immunoblotting (Snail1, (phospho)AMPK, (phospho)IκBα, (phospho)mTOR, cyclin D1, (phospho)Akt and ERα) and immunohistochemical analysis (E-cadherin). We have found that: 1) metformin treatment of MCF-7 cells is accompanied with the stimulation of AMPK and inhibition of growth-related proteins including IκBα, NF-κB, cyclin D1 and ERα; 2) long-term metformin treatment lead to the appearance and progression of cross-resistance to metformin and tamoxifen; the resistant cells are characterized with the unaffected AMPK activity, but the irreversible ER suppression and constitutive activation of Akt/Snail1 signaling; 3) Akt/Snail1 signaling is involved into progression of metformin resistance. The results presented may be considered as the first evidence of the progression of cross-resistance to metformin and tamoxifen in breast cancer cells. Importantly, the acquired resistance to both drugs is based on the constitutive activation of Akt/Snail1/E-cadherin signaling that opens new perspectives to overcome the metformin/tamoxifen resistance of breast cancer.
Collapse
Affiliation(s)
- Alexander M Scherbakov
- Laboratory of Clinical Biochemistry, Institute of Clinical Oncology, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Danila V Sorokin
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Victor V Tatarskiy
- Laboratory of Cell Death Mechanisms, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Nikolay S Prokhorov
- Laboratory of Microbial Viruses, S.N. Winogradsky Institute of Microbiology, Russian Academy of Sciences, Moscow, Russia
| | - Svetlana E Semina
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Lev M Berstein
- Laboratory of Oncoendocrinology, N.N. Petrov Research Institute of Oncology, St, Petersburg, Russia
| | - Mikhail A Krasil'nikov
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| |
Collapse
|
14
|
Brugnoli F, Grassilli S, Al-Qassab Y, Capitani S, Bertagnolo V. PLC-β2 is modulated by low oxygen availability in breast tumor cells and plays a phenotype dependent role in their hypoxia-related malignant potential. Mol Carcinog 2016; 55:2210-2221. [DOI: 10.1002/mc.22462] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/23/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Federica Brugnoli
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine; University of Ferrara; Ferrara Italy
| | - Silvia Grassilli
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine; University of Ferrara; Ferrara Italy
| | - Yasamin Al-Qassab
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine; University of Ferrara; Ferrara Italy
- Department of Anatomy, College of Medicine; University of Baghdad; Baghdad Iraq
| | - Silvano Capitani
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine; University of Ferrara; Ferrara Italy
- LTTA Centre; University of Ferrara; Ferrara Italy
| | - Valeria Bertagnolo
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine; University of Ferrara; Ferrara Italy
| |
Collapse
|
15
|
Gurzu S, Turdean S, Kovecsi A, Contac AO, Jung I. Epithelial-mesenchymal, mesenchymal-epithelial, and endothelial-mesenchymal transitions in malignant tumors: An update. World J Clin Cases 2015; 3:393-404. [PMID: 25984514 PMCID: PMC4419103 DOI: 10.12998/wjcc.v3.i5.393] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/12/2015] [Accepted: 04/02/2015] [Indexed: 02/05/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) represents conversion of an epithelial cell in an elongated cell with mesenchymal phenotype, which can occur in physiologic and pathologic processes such as embryogenesis (type 1 EMT), wound healing and/or fibrosis (type 2 EMT) and malignant tumors (type 3 EMT). The proliferation rate, metastasizing and recurrence capacity, as also the individualized response at chemotherapics, in both epithelial and mesenchymal malignant tumors is known to be influenced by reversible switch between EMT and mesenchymal-to-epithelial transition (MET). Although much research work has already been done in these fields, the specific molecular pathways of EMT, relating to the tumor type and tumor localization, are yet to be elucidated. In this paper, based on the literature and personal experience of the authors, an update in the field of EMT vs MET in epithelial and mesenchymal tumors is presented. The authors tried to present the latest data about the particularities of these processes, and also of the so-called endothelial-to-mesenchymal transition, based on tumor location. The EMT-angiogenesis link is discussed as a possible valuable parameter for clinical follow-up and targeted therapeutic oncologic management. The paper begins with presentation of the basic aspects of EMT, its classification and assessment possibilities, and concludes with prognostic and therapeutic perspectives. The particularities of EMT and MET in gastric and colorectal carcinomas, pancreatic cancer, hepatocellular and cholangiocarcinomas, and lung, breast and prostate cancers, respectively in sarcomas and gastrointestinal stromal tumors are presented in detail.
Collapse
|
16
|
Pilli VS, Gupta K, Kotha BP, Aradhyam GK. Snail-mediated Cripto-1 repression regulates the cell cycle and epithelial-mesenchymal transition-related gene expression. FEBS Lett 2015; 589:1249-56. [PMID: 25889638 DOI: 10.1016/j.febslet.2015.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/23/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
Transcription factor Snail mediates epithelial to mesenchymal transitions (EMT) by coordinate repression of epithelial markers, facilitating mass cell movement during germ layer formation. Aberrant reprogramming in its signaling pathways causes metastatic cancer. Snail's involvement in "fate-changing" decisions is however not understood. Cripto-1 shares a common temporal expression pattern with Snail during development. While Cripto-1 is required for mammary morphogenesis and hematopoietic stem cell renewal, its unregulated expression causes metastatic cancers. Therefore, we suspected that Snail regulates the expression of Cripto-1 controlling decisions such as motility, transformation and differentiation. We demonstrate that Snail represses Cripto-1 gene by direct transcriptional interaction and propose a novel mechanism by which it co-ordinately regulates cell fate decisions during development and could be causal of cancers.
Collapse
Affiliation(s)
- Vijaya S Pilli
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - K Gupta
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - Bhanu P Kotha
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - Gopala Krishna Aradhyam
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
17
|
Yang C, Li C, Li M, Tong X, Hu X, Yang X, Yan X, He L, Wan C. CYP2S1 depletion enhances colorectal cell proliferation is associated with PGE2-mediated activation of β-catenin signaling. Exp Cell Res 2014; 331:377-86. [PMID: 25557876 DOI: 10.1016/j.yexcr.2014.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 01/22/2023]
Abstract
Colorectal epithelial cancer is one of the most common cancers in the world and its 5-year survival rate is still relatively low. Cytochrome P450 (CYP) enzymes in epithelial cells lining the alimentary tract play an important role in the oxidative metabolism of a wide range of xenobiotics, including (pro-)carcinogens and endogenous compounds. Although CYP2S1, a member of CYP family, strongly expressed in many extrahepatic tissues, the role of CYP2S1 in cancer remains unclear. To investigate whether CYP2S1 involves in colorectal carcinogenesis, cell proliferation was analyzed in HCT116 cells depleted of CYP2S1 using small hairpin interfering RNA. Our data show that CYP2S1 knockdown promotes cell proliferation through increasing the level of endogenous prostaglandin E2(PGE2). PGE2, in turn, reduces phosphorylation of β-catenin and activates β-catenin signaling, which contributes to the cell proliferation. Furthermore, CYP2S1 knockdown increase tumor growth in xenograft mouse model. In brief, these results demonstrate that CYP2S1 regulates colorectal cancer growth through associated with PGE2-mediated activation of β-catenin signaling.
Collapse
Affiliation(s)
- Chao Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China; College of Life Science, Anhui Normal University, Wuhu 241000, Anhui, China
| | - Changyuan Li
- College of Life Science, Anhui Normal University, Wuhu 241000, Anhui, China
| | - Minle Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xuhan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaomei Yan
- School of Life Sciences & Biotechnology, Shanghai JiaoTong University, Shanghai 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
18
|
Kim MJ, Lim J, Yang Y, Lee MS, Lim JS. N-myc downstream-regulated gene 2 (NDRG2) suppresses the epithelial-mesenchymal transition (EMT) in breast cancer cells via STAT3/Snail signaling. Cancer Lett 2014; 354:33-42. [PMID: 25153349 DOI: 10.1016/j.canlet.2014.06.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/17/2014] [Accepted: 06/19/2014] [Indexed: 12/31/2022]
Abstract
Although NDRG2 has recently been found to be a candidate tumor suppressor, its precise role in the epithelial-mesenchymal transition (EMT) is not well understood. In the present study, we demonstrated that NDRG2 overexpression in MDA-MB-231 cells down-regulated the expression of Snail, a transcriptional repressor of E-cadherin and a key regulator of EMT, as well as the phosphorylation of signal transducer and activator of transcription 3 (STAT3), an oncogenic transcription factor that is activated in many human malignancies including breast cancer. In addition, we confirmed that the expression of Snail and phospho-STAT3 was recovered when NDRG2 was knocked down by siRNA in MCF7 cells in which NDRG2 is endogenously expressed. Interestingly, MDA-MB-231-NDRG2 cells showed remarkably decreased Snail expression after treatment with JSI-124 (also known as cucurbitacin I) or Stattic, STAT3 inhibitors, compared to MDA-MB-231-mock cells. Moreover, STAT3 activation by EGF treatment induced higher Snail expression, and NDRG2 overexpression resulted in the inhibition of Snail expression in MDA-MB-231 cells stimulated by EGF in the absence or presence of STAT3 inhibitor. Treatment of MDA-MB-231 cells with STAT3 inhibitor led to a moderate decrease in wound healing and migration capacity, whereas STAT3 inhibitor treatment of MDA-MB-231-NDRG2 cells resulted in a significant attenuation of migration in both resting and EGF-stimulated cells. Collectively, our data demonstrate that the inhibition of STAT3 signaling by NDRG2 suppresses EMT progression of EMT via the down-regulation of Snail expression.
Collapse
Affiliation(s)
- Myung-Jin Kim
- Department of Biological Science and the Research Center for Women's Diseases, Sookmyung Women's University, Hyochangwongil 52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Jihyun Lim
- Department of Biological Science and the Research Center for Women's Diseases, Sookmyung Women's University, Hyochangwongil 52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Young Yang
- Department of Biological Science and the Research Center for Women's Diseases, Sookmyung Women's University, Hyochangwongil 52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Myeong-Sok Lee
- Department of Biological Science and the Research Center for Women's Diseases, Sookmyung Women's University, Hyochangwongil 52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science and the Research Center for Women's Diseases, Sookmyung Women's University, Hyochangwongil 52, Yongsan-Gu, Seoul 140-742, Republic of Korea.
| |
Collapse
|
19
|
Wnt pathway activation increases hypoxia tolerance during development. PLoS One 2014; 9:e103292. [PMID: 25093834 PMCID: PMC4122365 DOI: 10.1371/journal.pone.0103292] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/27/2014] [Indexed: 11/19/2022] Open
Abstract
Adaptation to hypoxia, defined as a condition of inadequate oxygen supply, has enabled humans to successfully colonize high altitude regions. The mechanisms attempted by organisms to cope with short-term hypoxia include increased ATP production via anaerobic respiration and stabilization of Hypoxia Inducible Factor 1α (HIF-1α). However, less is known about the means through which populations adapt to chronic hypoxia during the process of development within a life time or over generations. Here we show that signaling via the highly conserved Wnt pathway impacts the ability of Drosophila melanogaster to complete its life cycle under hypoxia. We identify this pathway through analyses of genome sequencing and gene expression of a Drosophila melanogaster population adapted over >180 generations to tolerate a concentration of 3.5-4% O2 in air. We then show that genetic activation of the Wnt canonical pathway leads to increased rates of adult eclosion in low O2. Our results indicate that a previously unsuspected major developmental pathway, Wnt, plays a significant role in hypoxia tolerance.
Collapse
|