1
|
Tong X, Burks HE, Ren Z, Koetsier JL, Roth-Carter QR, Green KJ. Crosstalk in Skin: Loss of Desmoglein 1 in Keratinocytes Inhibits BRAF V600E-Induced Cellular Senescence in Human Melanocytes. J Invest Dermatol 2024:S0022-202X(24)02955-5. [PMID: 39581457 DOI: 10.1016/j.jid.2024.10.608] [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: 01/19/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024]
Abstract
Melanoma arises from transformation of melanocytes in the basal layer of epidermis where they are surrounded by keratinocytes, with which they interact through cell contact and paracrine communication. Although research focuses on how the accumulation of oncogene and tumor suppressor gene mutations in melanocytes drive melanomagenesis, how alterations in keratinocytes serve as extrinsic drivers of melanoma initiation and progression is poorly understood. We recently identified keratinocyte desmoglein 1 (DSG1) as an mediator of keratinocyte:melanoma crosstalk. In this study, we address the extent to which DSG1 loss, which occurs in response to environmental stress such as UVR, affects early steps in melanomagenesis. RNA-sequencing analysis revealed that paracrine signals from DSG1-deficient keratinocytes mediate a transcriptional switch from a differentiated to undifferentiated cell state in melanocytes expressing BRAFV600E. Of 221 differentially expressed genes in BRAFV600E cells treated with conditioned media from DSG1-deficient keratinocytes, the laminin superfamily member Netrin-4 (NTN4), which inhibits senescence, stood out. Indeed, although BRAFV600E melanocytes treated with conditioned media from DSG1-deficient keratinocytes showed signs of senescence bypass, NTN4 knockdown reversed these effects, whereas ectopic Netrin-4 expression mimicked them. These results suggest that DSG1 loss in keratinocytes provides an extrinsic signal to push melanocytes toward oncogenic transformation once an initial mutation has been introduced.
Collapse
Affiliation(s)
- Xin Tong
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Hope E Burks
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jennifer L Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Quinn R Roth-Carter
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
2
|
Green KJ, Pokorny J, Jarrell B. Dangerous liaisons: Loss of keratinocyte control over melanocytes in melanomagenesis. Bioessays 2024; 46:e2400135. [PMID: 39233509 PMCID: PMC11626500 DOI: 10.1002/bies.202400135] [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: 06/03/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
Melanomas arise from transformed melanocytes, positioned at the dermal-epidermal junction in the basal layer of the epidermis. Melanocytes are completely surrounded by keratinocyte neighbors, with which they communicate through direct contact and paracrine signaling to maintain normal growth control and homeostasis. UV radiation from sunlight reshapes this communication network to drive a protective tanning response. However, repeated rounds of sun exposure result in accumulation of mutations in melanocytes that have been considered as primary drivers of melanoma initiation and progression. It is now clear that mutations in melanocytes are not sufficient to drive tumor formation-the tumor environment plays a critical role. This review focuses on changes in melanocyte-keratinocyte communication that contribute to melanoma initiation and progression, with a particular focus on recent mechanistic insights that lay a foundation for developing new ways to intercept melanoma development.
Collapse
Affiliation(s)
- Kathleen J. Green
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoUSA
- Department of DermatologyNorthwestern University Feinberg School of MedicineChicagoUSA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoUSA
| | - Jenny Pokorny
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoUSA
| | - Brieanna Jarrell
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoUSA
| |
Collapse
|
3
|
Gao X, Ye J, Huang X, Huang S, Luo W, Zeng D, Li S, Tang M, Mai R, Li Y, Lin Y, Liang R. Research progress of the netrins and their receptors in cancer. J Cell Mol Med 2024; 28:e18241. [PMID: 38546656 PMCID: PMC10977403 DOI: 10.1111/jcmm.18241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 11/12/2024] Open
Abstract
Netrins, a family of secreted and membrane-associated proteins, can regulate axonal guidance, morphogenesis, angiogenesis, cell migration, cell survival, and tumorigenesis. Four secreted netrins (netrin 1, 3, 4 and 5) and two glycosylphosphatidylinositols-anchored membrane proteins, netrin-G1 and G2, have been identified in mammals. Netrins and their receptors can serve as a biomarker and molecular therapeutic target for pathological differentiation, diagnosis and prognosis of malignant cancers. We review here the potential roles of the netrins family and their receptors in cancer.
Collapse
Affiliation(s)
- Xing Gao
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Jiazhou Ye
- Department of Hepatobiliary SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Xi Huang
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Shilin Huang
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Wenfeng Luo
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Dandan Zeng
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Shizhou Li
- Department of Hepatobiliary SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Minchao Tang
- Department of Hepatobiliary SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Rongyun Mai
- Department of Hepatobiliary SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Yongqiang Li
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Yan Lin
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Rong Liang
- Department of Digestive OncologyGuangxi Medical University Cancer HospitalNanningGuangxiChina
| |
Collapse
|
4
|
Shi YH, Li JQ, Min-Xu, Wang YY, Wang TH, Zuo ZF, Liu XZ. Bioinformatics-based Study on the Effects of Umbilical Cord Mesenchymal Stem Cells on the Aging Retina. Curr Stem Cell Res Ther 2024; 19:1497-1513. [PMID: 38204243 DOI: 10.2174/011574888x277276231215110316] [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: 09/02/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Retinal aging is one of the common public health problems caused by population aging and has become an important cause of acquired vision loss in adults. The aim of this study was to determine the role of human umbilical cord mesenchymal stem cells (hUCMSCs) in delaying retinal ganglion cell (RGC) aging and part of the network of molecular mechanisms involved. METHODS A retinal ganglion cell senescence model was established in vitro and treated with UCMSC. Successful establishment of the senescence system was demonstrated using β- galactosidase staining. The ameliorative effect of MSC on senescence was demonstrated using CCK8 cell viability and Annexin V-PI apoptosis staining. The relevant targets of RGC, MSC, and senescence were mainly obtained by searching the GeneCards database. The protein interaction network among the relevant targets was constructed using the String database and Cytoscape, and 10 key target genes were calculated based on the MCC algorithm, based on which Gene ontologies (GO) enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were performed. Changes in relevant target genes were detected using real-time fluorescence quantitative PCR and the mechanism of action of UCMSC was determined by RNA interference. RESULTS β-galactosidase staining showed that UCMSC significantly reduced the positive results of RGC. The retinal aging process was alleviated. The bioinformatics screen yielded 201 shared genes. 10 key genes were selected by the MCC algorithm, including vascular endothelial growth factor A (VEGFA), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), albumin (ALB), interleukin- 6 (IL6), tumor necrosis factor (TNF), tumor protein P53 (TP53), insulin (INS), matrix metalloproteinase 9 (MMP9), epidermal growth factor (EGF), interleukin-1β (IL1B), and enrichment to related transferase activity and kinase activity regulated biological processes involved in oxidative stress and inflammation related pathways. In addition, PCR results showed that all the above molecules were altered in expression after UCMSC involvement. CONCLUSION This experiment demonstrated the role of UCMSC in delaying retinal ganglion cell senescence and further elucidated that UCMSC may be associated with the activation of VEGFA, TP53, ALB, GAPDH, IL6, IL1B, MMP9 genes and the inhibition of INS, EGF, and TNF in delaying retinal senescence.
Collapse
Affiliation(s)
- Ya-Hui Shi
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
| | - Jun-Qi Li
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
| | - Min-Xu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
| | - Yu-Ying Wang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
| | - Ting-Hua Wang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, China
| | - Zhong-Fu Zuo
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, China
| | - Xue-Zheng Liu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, 121000, China
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, China
| |
Collapse
|
5
|
Shah MA, Abuzar SM, Ilyas K, Qadees I, Bilal M, Yousaf R, Kassim RMT, Rasul A, Saleem U, Alves MS, Khan H, Blundell R, Jeandet P. Ginsenosides in cancer: Targeting cell cycle arrest and apoptosis. Chem Biol Interact 2023; 382:110634. [PMID: 37451663 DOI: 10.1016/j.cbi.2023.110634] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Despite the existence of extensive clinical research and novel therapeutic treatments, cancer remains undefeated and the significant cause of death worldwide. Cancer is a disease in which growth of cells goes out of control, being also able to invade other parts of the body. Cellular division is strictly controlled by multiple checkpoints like G1/S and G2/M which, when dysregulated, lead to uncontrollable cell division. The current remedies which are being utilized to combat cancer are monoclonal antibodies, chemotherapy, cryoablation, and bone marrow transplant etc. and these have also been greatly disheartening because of their serious adverse effects like hypotension, neuropathy, necrosis, leukemia relapse and many more. Bioactive compounds derived from natural products have marked the history of the development of novel drug therapies against cancer among which ginsenosides have no peer as they target several signaling pathways, which when abnormally regulated, lead to cancer. Substantial research has reported that ginsenosides like Rb1, Rb2, Rb3, Rc, Rd, Rg3, Rh2 etc. can prevent and treat cancer by targeting different pathways and molecules by induction of autophagy, neutralizing ROS, induction of cancerous cell death by controlling the p53 pathway, modulation of miRNAs by decreasing Smad2 expression, regulating Bcl-2 expression by normalizing the NF-Kb pathway, inhibition of inflammatory pathways by decreasing the production of cytokines like IL-8, causing cell cycle arrest by restricting cyclin E1 and CDC2, and induction of apoptosis during malignancy by decreasing β-catenin levels etc. In this review, we have analyzed the anti-cancer therapeutic potential of various ginsenoside compounds in order to consider their possible use in new strategies in the fight against cancer.
Collapse
Affiliation(s)
| | - Syed Muhammad Abuzar
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kainat Ilyas
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Irtaza Qadees
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Momna Bilal
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Rimsha Yousaf
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | | | - Azhar Rasul
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Maria Silvana Alves
- Laboratory of Cellular and Molecular Bioactivity, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Renald Blundell
- Department of Physiology and Biochemistry, Faculty of Medicine, University of Malta, Msida, MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, MSD2080 Imsida, Malta
| | - Philippe Jeandet
- University of Reims, Research Unit Induced Resistance and Plant Bioprotection USC INRAe 1488 Department of Biology and Biochemistry, Faculty of Sciences, 51100, Reims, France.
| |
Collapse
|
6
|
Liu A, Wang X, Hu L, Yan D, Yin Y, Zheng H, Liu G, Zhang J, Li Y. A predictive molecular signature consisting of lncRNAs associated with cellular senescence for the prognosis of lung adenocarcinoma. PLoS One 2023; 18:e0287132. [PMID: 37352167 PMCID: PMC10289466 DOI: 10.1371/journal.pone.0287132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/31/2023] [Indexed: 06/25/2023] Open
Abstract
The role of long noncoding RNAs (lncRNAs) has been verified by more and more researches in recent years. However, there are few reports on cellular senescence-associated lncRNAs in lung adenocarcinoma (LUAD). Therefore, to explore the prognostic effect of lncRNAs in LUAD, 279 cellular senescence-related genes, survival information and clinicopathologic parameters were derived from the CellAge database and The Cancer Genome Atlas (TCGA) database. Then, we constructed a novel cellular senescence-associated lncRNAs predictive signature (CS-ALPS) consisting of 6 lncRNAS (AC026355.1, AL365181.2, AF131215.5, C20orf197, GAS6-AS1, GSEC). According to the median of the risk score, 480 samples were divided into high-risk and low-risk groups. Furthermore, the clinicopathological and biological functions, immune characteristics and common drug sensitivity were analyzed between two risk groups. In conclusion, the CS-ALPS can independently forecast the prognosis of LUAD, which reveals the potential molecular mechanism of cellular senescence-associated lncRNAs, and provides appropriate strategies for the clinical treatment of patients with LUAD.
Collapse
Affiliation(s)
- Anbang Liu
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaohuai Wang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liu Hu
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dongqing Yan
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yin Yin
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hongjie Zheng
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Gengqiu Liu
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Junhang Zhang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yun Li
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
7
|
Mu J, Gong J, Shi M, Zhang Y. Analysis and validation of aging-related genes in prognosis and immune function of glioblastoma. BMC Med Genomics 2023; 16:109. [PMID: 37208656 DOI: 10.1186/s12920-023-01538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 05/09/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a common malignant brain tumor with poor prognosis and high mortality. Numerous reports have identified the correlation between aging and the prognosis of patients with GBM. The purpose of this study was to establish a prognostic model for GBM patients based on aging-related gene (ARG) to help determine the prognosis of GBM patients. METHODS 143 patients with GBM from The Cancer Genomic Atlas (TCGA), 218 patients with GBM from the Chinese Glioma Genomic Atlas (CGGA) of China and 50 patients from Gene Expression Omnibus (GEO) were included in the study. R software (V4.2.1) and bioinformatics statistical methods were used to develop prognostic models and study immune infiltration and mutation characteristics. RESULTS Thirteen genes were screened out and used to establish the prognostic model finally, and the risk scores of the prognostic model was an independent factor (P < 0.001), which indicated a good prediction ability. In addition, there are significant differences in immune infiltration and mutation characteristics between the two groups with high and low risk scores. CONCLUSION The prognostic model of GBM patients based on ARGs can predict the prognosis of GBM patients. However, this signature requires further investigation and validation in larger cohort studies.
Collapse
Affiliation(s)
- Jianhua Mu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jianan Gong
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Miao Shi
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Yinian Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Tong X, Burks HE, Ren Z, Koetsier JL, Roth-Carter QR, Green KJ. Crosstalk in skin: Loss of desmoglein 1 in keratinocytes inhibits BRAF V600E-induced cellular senescence in human melanocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528886. [PMID: 36824910 PMCID: PMC9949056 DOI: 10.1101/2023.02.16.528886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Melanoma arises from transformation of melanocytes in the basal layer of the epidermis where they are surrounded by keratinocytes, with which they interact through cell contact and paracrine communication. Considerable effort has been devoted to determining how the accumulation of oncogene and tumor suppressor gene mutations in melanocytes drive melanoma development. However, the extent to which alterations in keratinocytes that occur in the developing tumor niche serve as extrinsic drivers of melanoma initiation and progression is poorly understood. We recently identified the keratinocyte-specific cadherin, desmoglein 1 (Dsg1), as an important mediator of keratinocyte:melanoma cell crosstalk, demonstrating that its chronic loss, which can occur through melanoma cell-dependent paracrine signaling, promotes behaviors that mimic a malignant phenotype. Here we address the extent to which Dsg1 loss affects early steps in melanomagenesis. RNA-Seq analysis revealed that paracrine signals from Dsg1-deficient keratinocytes mediate a transcriptional switch from a differentiated to undifferentiated cell state in melanocytes expressing BRAFV600E, a driver mutation commonly present in both melanoma and benign nevi and reported to cause growth arrest and oncogene-induced senescence (OIS). Of ~220 differentially expressed genes in BRAFV600E cells treated with Dsg1-deficient conditioned media (CM), the laminin superfamily member NTN4/Netrin-4, which inhibits senescence in endothelial cells, stood out. Indeed, while BRAFV600E melanocytes treated with Dsg1-deficient CM showed signs of senescence bypass as assessed by increased senescence-associated β-galactosidase activity and decreased p16, knockdown of NTN4 reversed these effects. These results suggest that Dsg1 loss in keratinocytes provides an extrinsic signal to push melanocytes towards oncogenic transformation once an initial mutation has been introduced.
Collapse
Affiliation(s)
- Xin Tong
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Hope E. Burks
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jennifer L. Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Quinn R. Roth-Carter
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kathleen J. Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
9
|
Ma F, Ma Y, Liu K, Gao J, Li S, Sun X, Li G. Resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis. Food Funct 2023; 14:1484-1497. [PMID: 36651299 DOI: 10.1039/d2fo01188c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inducing cell senescence is widely regarded as a potent tumor suppression mechanism. Resveratrol has attracted increasing attention for its capacity to prevent and suppress cancer. However, the mechanism of resveratrol on the induction of cancer cell senescence has not been well clarified. Our results showed that resveratrol inhibited cell viability and colony formation and promoted cell senescence along with augmentation of SA-β-gal activity and modulation of senescence-associated molecular markers p53, p21 and LaminB protein in breast and liver cancer cells. The underlying mechanism was that resveratrol increased ROS generation to enhance tumor suppressor gene DLC1 expression, and DLC1 further inhibited the DYRK1A-EGFR axis to trigger DNA damage accompanied by up-regulation of the DNA double strand break marker protein γH2AX and down-regulation of the DNA repair related proteins p-BRCA1 and RAD51, eventually leading to cancer cell senescence. Resveratrol also effectively inhibited the volume of transplanted tumor with increased SA-β-gal activity and DLC1 level in a chicken embryo allantoic membrane xenograft tumor model. This is the first report to investigate whether resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis, which could provide a solid base for resveratrol's application in cancer prevention and clinical treatment as a food additive or adjuvant therapies.
Collapse
Affiliation(s)
- Fengqiu Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Keke Liu
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Shasha Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Xiaowen Sun
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| |
Collapse
|
10
|
Dong F, Liu Y, Yan W, Meng Q, Song X, Cheng B, Yao R. Netrin-4: Focus on Its Role in Axon Guidance, Tissue Stability, Angiogenesis and Tumors. Cell Mol Neurobiol 2022:10.1007/s10571-022-01279-4. [DOI: 10.1007/s10571-022-01279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/26/2022] [Indexed: 11/11/2022]
|
11
|
Gong L, Yin Y, Chen C, Wan Q, Xia D, Wang M, Pu Z, Zhang B, Zou J. Characterization of EGFR-reprogrammable temozolomide-resistant cells in a model of glioblastoma. Cell Death Dis 2022; 8:438. [PMID: 36316307 PMCID: PMC9622861 DOI: 10.1038/s41420-022-01230-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2022]
Abstract
Temozolomide (TMZ) resistance is a major clinical challenge for glioblastoma (GBM). O6-methylguanine-DNA methyltransferase (MGMT) mediated DNA damage repair is a key mechanism for TMZ resistance. However, MGMT-null GBM patients remain resistant to TMZ, and the process for resistance evolution is largely unknown. Here, we developed an acquired TMZ resistant xenograft model using serial implantation of MGMT-hypermethylated U87 cells, allowing the extraction of stable, TMZ resistant (TMZ-R) tumors and primary cells. The derived tumors and cells exhibited stable multidrug resistance both in vitro and in vivo. Functional experiments, as well as single-cell RNA sequencing (scRNA-seq), indicated that TMZ treatment induced cellular heterogeneity including quiescent cancer stem cells (CSCs) in TMZ-R tumors. A subset of these were labeled by NES+/SOX2+/CADM1+ and demonstrated significant advantages for drug resistance. Further study revealed that Epidermal Growth Factor Receptor (EGFR) deficiency and diminished downstream signaling may confer this triple positive CSCs subgroup’s quiescent phenotypes and chemoresistance. Continuous EGF treatment improved the chemosensitivity of TMZ-R cells both in vitro and in vivo, mechanically reversing cell cycle arrest and reduced drug uptake. Further, EGF treatment of TMZ-R tumors favorably normalized the response to TMZ in combination therapy. Here, we characterize a unique subgroup of CSCs in MGMT-null experimental glioblastoma, identifying EGF + TMZ therapy as a potential strategy to overcome cellular quiescence and TMZ resistance, likely endowed by deficient EGFR signaling.
Collapse
Affiliation(s)
- Lingli Gong
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Ying Yin
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Cheng Chen
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Quan Wan
- grid.89957.3a0000 0000 9255 8984Department of Neurosurgery, The Affiliated Wuxi Second Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002 China
| | - Die Xia
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Mei Wang
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Zhening Pu
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Bo Zhang
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| | - Jian Zou
- grid.89957.3a0000 0000 9255 8984Department of Laboratory Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China ,grid.89957.3a0000 0000 9255 8984Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, 214023 China
| |
Collapse
|
12
|
Yang J, Liu M, Hong D, Zeng M, Zhang X. The Paradoxical Role of Cellular Senescence in Cancer. Front Cell Dev Biol 2021; 9:722205. [PMID: 34458273 PMCID: PMC8388842 DOI: 10.3389/fcell.2021.722205] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence occurs in proliferating cells as a consequence of various triggers including telomere shortening, DNA damage, and inappropriate expression of oncogenes. The senescent state is accompanied by failure to reenter the cell cycle under mitotic stimulation, resistance to cell death and enhanced secretory phenotype. A growing number of studies have convincingly demonstrated a paradoxical role for spontaneous senescence and therapy-induced senescence (TIS), that senescence may involve both cancer prevention and cancer aggressiveness. Cellular senescence was initially described as a physiological suppressor mechanism of tumor cells, because cancer development requires cell proliferation. However, there is growing evidence that senescent cells may contribute to oncogenesis, partly in a senescence-associated secretory phenotype (SASP)-dependent manner. On the one hand, SASP prevents cell division and promotes immune clearance of damaged cells, thereby avoiding tumor development. On the other hand, SASP contributes to tumor progression and relapse through creating an immunosuppressive environment. In this review, we performed a review to summarize both bright and dark sides of senescence in cancer, and the strategies to handle senescence in cancer therapy were also discussed.
Collapse
Affiliation(s)
- Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dongchun Hong
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
13
|
Xia Q, Liu L, Li Y, Zhang P, Han D, Dong L. Therapeutic Perspective of Temozolomide Resistance in Glioblastoma Treatment. Cancer Invest 2021; 39:627-644. [PMID: 34254870 DOI: 10.1080/07357907.2021.1952595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glioblastoma (GB) is the most lethal form of primary brain neoplasm. TMZ is the first-line standard treatment, but the strong resistance constrains the efficacy in clinical use. GB contains glioma stem cells (GSCs), which contribute to TMZ resistance, promote cell survival evolvement, and repopulate the tumor mass. This review summarizes the TMZ-resistance mechanisms and discusses several potential therapies from the conservative opinion of GSC-targeted therapy orientation to the current view of TMZ resistance-aimed efficacy, which will provide an understanding of the role of heterogeneity in drug resistance and improve therapeutic efficacy in general.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Pei Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Han
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
14
|
Zhu YG, Lv YX, Guo CY, Xiao ZM, Jiang QG, Kuang H, Zhang WH, Hu P. Harmine inhibits the proliferation and migration of glioblastoma cells via the FAK/AKT pathway. Life Sci 2021; 270:119112. [DOI: 10.1016/j.lfs.2021.119112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 01/01/2023]
|
15
|
HIF1α/HIF2α-Sox2/Klf4 promotes the malignant progression of glioblastoma via the EGFR-PI3K/AKT signalling pathway with positive feedback under hypoxia. Cell Death Dis 2021; 12:312. [PMID: 33762574 PMCID: PMC7990922 DOI: 10.1038/s41419-021-03598-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Previous studies have suggested that hypoxic responses are regulated by hypoxia-inducible factors (HIFs), which in turn promote the malignant progression of glioblastoma (GBM) by inhibiting apoptosis and increasing proliferation; these events lead to a poor prognosis of GBM patients. However, there are still no HIF-targeted therapies for the treatment of GBM. We have conducted series of experiments and discovered that GBM cells exhibit features indicative of malignant progression and are present in a hypoxic environment. Knocking out HIF1α or HIF2α alone resulted in no significant change in cell proliferation and cell cycle progression in response to acute hypoxia, but cells showed inhibition of stemness expression and chemosensitization to temozolomide (TMZ) treatment. However, simultaneously knocking out HIF1α and HIF2α inhibited cell cycle arrest and promoted proliferation with decreased stemness, making GBM cells more sensitive to chemotherapy, which could improve patient prognosis. Thus, HIF1α and HIF2α regulate each other with negative feedback. In addition, HIF1α and HIF2α are upstream regulators of epidermal growth factor (EGF), which controls the malignant development of GBM through the EGFR-PI3K/AKT-mTOR-HIF1α signalling pathway. In brief, the HIF1α/HIF2α-EGF/EGFR-PI3K/AKT-mTOR-HIF1α signalling axis contributes to the growth of GBM through a positive feedback mechanism. Finally, HIF1α and HIF2α regulate Sox2 and Klf4, contributing to stemness expression and inducing cell cycle arrest, thus increasing malignancy in GBM. In summary, HIF1α and HIF2α regulate glioblastoma malignant progression through the EGFR-PI3K/AKT pathway via a positive feedback mechanism under the effects of Sox2 and Klf4, which provides a new tumour development model and strategy for glioblastoma treatment.
Collapse
|
16
|
Xu Q, Hu C, Zhu Y, Wang K, Lal B, Li L, Tang J, Wei S, Huang G, Xia S, Lv S, Laterra J, Jiang Y, Li Y. ShRNA-based POLD2 expression knockdown sensitizes glioblastoma to DNA-Damaging therapeutics. Cancer Lett 2020; 482:126-135. [PMID: 31954770 DOI: 10.1016/j.canlet.2020.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
Abstract
Glioblastoma (GBM) has limited therapeutic options. DNA repair mechanisms contribute GBM cells to escape therapies and re-establish tumor growth. Multiple studies have shown that POLD2 plays a critical role in DNA replication, DNA repair and genomic stability. We demonstrate for the first time that POLD2 is highly expressed in human glioma specimens and that expression correlates with poor patient survival. siRNA or shRNA POLD2 inhibited GBM cell proliferation, cell cycle progression, invasiveness, sensitized GBM cells to chemo/radiation-induced cell death and reversed the cytoprotective effects of EGFR signaling. Conversely, forced POLD2 expression was found to induce GBM cell proliferation, colony formation, invasiveness and chemo/radiation resistance. POLD2 expression associated with stem-like cell subsets (CD133+ and SSEA-1+ cells) and positively correlated with Sox2 expression in clinical specimens. Its expression was induced by Sox2 and inhibited by the forced differentiation of GBM neurospheres. shRNA-POLD2 modestly inhibited GBM neurosphere-derived orthotopic xenografts growth, when combined with radiation, dramatically inhibited xenograft growth in a cooperative fashion. These novel findings identify POLD2 as a new potential therapeutic target for enhancing GBM response to current standard of care therapeutics.
Collapse
Affiliation(s)
- Qingfu Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, PR China; Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China; Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Chengchen Hu
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Yan Zhu
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, PR China; Department of Obstetrics and Gynecology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Kimberly Wang
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA
| | - Bachuchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA
| | - Lichao Li
- Department of Neurosurgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Junhai Tang
- Department of Neurosurgery, Third Military Medical University, Chongqing, 400037, PR China
| | - Shuang Wei
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA
| | - Guohao Huang
- Department of Neurosurgery, Third Military Medical University, Chongqing, 400037, PR China
| | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Shengqing Lv
- Department of Neurosurgery, Third Military Medical University, Chongqing, 400037, PR China
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Neuroscience, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N. Broadway, Baltimore, MD, 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
17
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Tian ZR, Sahib S, Bryukhovetskiy I, Bryukhovetskiy A, Buzoianu AD, Patnaik R, Wiklund L, Sharma A. Pathophysiology of blood-brain barrier in brain tumor. Novel therapeutic advances using nanomedicine. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:1-66. [PMID: 32448602 DOI: 10.1016/bs.irn.2020.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|