1
|
Fahim SA, El Sobky SA, Abdellatif A, Fawzy IO, Abdelaziz AI. MEIS1: From functional versatility to post-transcriptional/translational regulation. Life Sci 2025; 374:123683. [PMID: 40339957 DOI: 10.1016/j.lfs.2025.123683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is a transcription factor involved in a myriad of functions such as hematopoiesis, cardiac regeneration, cell cycle progression, and limb and organ development. Its functional versatility extends beyond developmental biology, as aberrant MEIS1 expression has been implicated in various pathological contexts like carcinogenesis, cardiomyopathies, and neurodegenerative disorders. Recent advances in the field have uncovered novel layers of MEIS1 regulation, focusing on post-transcriptional and translational mechanisms, which collectively fine-tune its activity, stability, and subcellular localization. These include chromatin remodeling, epigenetic modifications in the enhancer and promoter regions, and protein modifications like phosphorylation and ubiquitination. The sophisticated regulation of MEIS1 including its interplay with non-coding RNAs (ncRNAs), either being an upstream or downstream of ncRNAs, equally represents an important regulatory mechanism orchestrating MEIS1 expression and function. This review explores the multifaceted roles of MEIS1, emphasizing its dynamic regulatory networks and their implications in physiological and pathological conditions. It also provides forward-thinking guidance on the utilization of MEIS1 in targeted therapies across various clinical settings, highlighting its potential as a key regulatory factor in disease modulation and therapeutic innovation.
Collapse
Affiliation(s)
- Salma A Fahim
- School of Medicine, Newgiza University (NGU), Giza, Egypt; Biotechnology Program, American University in Cairo, New Cairo, Egypt
| | | | - Ahmed Abdellatif
- Biotechnology Program, American University in Cairo, New Cairo, Egypt
| | | | | |
Collapse
|
2
|
Appunni S, Saxena A, Ramamoorthy V, Zhang Y, Doke M, Nair SS, Khosla AA, Rubens M. Decorin: matrix-based pan-cancer tumor suppressor. Mol Cell Biochem 2025; 480:3569-3591. [PMID: 39954173 DOI: 10.1007/s11010-025-05224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Studies have shown that decorin is a potent pan-cancer tumor suppressor that is under-expressed in most cancers. Decorin interacts with receptor tyrosine kinases and functions as a pan-receptor tyrosine kinase inhibitor, thereby suppressing oncogenic signals. Decorin deficiency promotes epithelial-to-mesenchymal transition and enhances cancer dissemination and metastasis. According to recent GLOBOCAN estimates, the most common cancers worldwide are breast, lung, prostate, colorectal, skin (non-melanoma), and stomach. Considering the burden of rising cancer incidence and the importance of discovering novel molecular markers and potential therapeutic agents for cancer management, we have outlined the possible expressional and clinicopathological significance of decorin in major cancers based on available pre-clinical and clinical studies. Measuring plasma decorin is a minimally invasive technique, and human studies have shown that it is useful in predicting clinical outcomes in cancer though it needs further validation. Oncolytic virus-mediated decorin gene transfer has shown significant anti-tumorigenic effects in pre-clinical studies, though its implication in human subjects is yet to be understood. Exogenous decorin delivery in experimental studies has been shown to mitigate cancer growth, but its therapeutic efficacy and safety are poorly understood. Future research is required to translate the tumor-suppressive action of decorin observed in preclinical experiments to therapeutic interventions in human subjects.
Collapse
Affiliation(s)
| | - Anshul Saxena
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St, Miami, FL, 33199, USA
- Baptist Health South Florida, Miami, FL, 33176, USA
| | | | - Yanjia Zhang
- Baptist Health South Florida, Miami, FL, 33176, USA
| | - Mayur Doke
- Miller School of Medicine, University of Miami, Coral Gables, FL, 33146, USA
| | - Sudheesh S Nair
- School of Veterinary Medicine, Ross University, Basseterre, Saint Kitts and Nevis
| | | | - Muni Rubens
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St, Miami, FL, 33199, USA.
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 33172, USA.
- Universidad Espíritu Santo, Samborondón, Ecuador.
| |
Collapse
|
3
|
Tabibian M, Moghaddam FS, Motevaseli E, Ghafouri-Fard S. Targeting mRNA-coding genes in prostate cancer using CRISPR/Cas9 technology with a special focus on androgen receptor signaling. Cell Commun Signal 2024; 22:504. [PMID: 39420406 PMCID: PMC11484332 DOI: 10.1186/s12964-024-01833-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Prostate cancer is among prevalent cancers in men. Numerous strategies have been proposed to intervene with the important prostate cancer-related signaling pathways. Among the most promising strategies is CRISPR/Cas9 strategy. This strategy has been used to modify expression of a number of genes in prostate cancer cells. AIMS This review summarizes the most recent progresses in the application of CRISPR/Cas9 strategy in modification of prostate cancer-related phenotypes with an especial focus on pathways related to androgen receptor signaling. CONCLUSION CRISPR/Cas9 technology has successfully targeted several genes in the prostate cancer cells. Moreover, the efficiency of this technique in reducing tumor burden has been tested in animal models of prostate cancer. Most of targeted genes have been related with the androgen receptor signaling. Targeted modulation of these genes have affected growth of castration-resistant prostate cancer. PI3K/AKT/mTOR signaling and immune response-related genes have been other targets that have been successfully modulated by CRISPR/Cas9 technology in prostate cancer. Based on the rapid translation of this technology into the clinical application, it is anticipated that novel treatments based on this technique change the outcome of this malignancy in future.
Collapse
Affiliation(s)
- Mobina Tabibian
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnologies, Shahid Beheshti University, Tehran, Iran
| | | | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Rahman M, Akter K, Ahmed KR, Fahim MMH, Aktary N, Park MN, Shin SW, Kim B. Synergistic Strategies for Castration-Resistant Prostate Cancer: Targeting AR-V7, Exploring Natural Compounds, and Optimizing FDA-Approved Therapies. Cancers (Basel) 2024; 16:2777. [PMID: 39199550 PMCID: PMC11352813 DOI: 10.3390/cancers16162777] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) remains a significant therapeutic challenge due to its resistance to standard androgen deprivation therapy (ADT). The emergence of androgen receptor splice variant 7 (AR-V7) has been implicated in CRPC progression, contributing to treatment resistance. Current treatments, including first-generation chemotherapy, androgen receptor blockers, radiation therapy, immune therapy, and PARP inhibitors, often come with substantial side effects and limited efficacy. Natural compounds, particularly those derived from herbal medicine, have garnered increasing interest as adjunctive therapeutic agents against CRPC. This review explores the role of AR-V7 in CRPC and highlights the promising benefits of natural compounds as complementary treatments to conventional drugs in reducing CRPC and overcoming therapeutic resistance. We delve into the mechanisms of action underlying the anti-CRPC effects of natural compounds, showcasing their potential to enhance therapeutic outcomes while mitigating the side effects associated with conventional therapies. The exploration of natural compounds offers promising avenues for developing novel treatment strategies that enhance therapeutic outcomes and reduce the adverse effects of conventional CRPC therapies. These compounds provide a safer, more effective approach to managing CRPC, representing a significant advancement in improving patient care.
Collapse
Affiliation(s)
- Muntajin Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Khadija Akter
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| | - Kazi Rejvee Ahmed
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Md. Maharub Hossain Fahim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Nahida Aktary
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Sang-Won Shin
- Department of Humanities & Social Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si 50612, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| |
Collapse
|
5
|
Kim HJ, Batara DC, Jeon YJ, Lee S, Beck S, Kim SH. The impact of MEIS1 TALE homeodomain transcription factor knockdown on glioma stem cell growth. Anim Cells Syst (Seoul) 2024; 28:93-109. [PMID: 38487309 PMCID: PMC10939110 DOI: 10.1080/19768354.2024.2327340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is a HOX co-factor necessary for organ development and normal hematopoiesis. Recently, MEIS1 has been linked to the development and progression of various cancers. However, its role in gliomagenesis particularly on glioma stem cells (GSCs) remains unclear. Here, we demonstrate that MEIS1 is highly upregulated in GSCs compared to normal, and glioma cells and to its differentiated counterparts. Inhibition of MEIS1 expression by shRNA significantly reduced GSC growth in both in vitro and in vivo experiments. On the other hand, integrated transcriptomics analyses of glioma datasets revealed that MEIS1 expression is correlated to cell cycle-related genes. Clinical data analysis revealed that MEIS1 expression is elevated in high-grade gliomas, and patients with high MEIS1 levels have poorer overall survival outcomes. The findings suggest that MEIS1 is a prognostic biomarker for glioma patients and a possible target for developing novel therapeutic strategies against GBM.
Collapse
Affiliation(s)
- Hyun-Jin Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Don Carlo Batara
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Samuel Beck
- Department of Dermatology, Center for Aging Research, Chobanian & Avedisian School of Medicine, Boston University, Boston, USA
| | - Sung-Hak Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
6
|
Abstract
In this comprehensive review, we will dissect the impact of research on proteoglycans focusing on recent developments involved in their synthesis, degradation, and interactions, while critically assessing their usefulness in various biological processes. The emerging roles of proteoglycans in global infections, specifically the SARS-CoV-2 pandemic, and their rising functions in regenerative medicine and biomaterial science have significantly affected our current view of proteoglycans and related compounds. The roles of proteoglycans in cancer biology and their potential use as a next-generation protein-based adjuvant therapy to combat cancer is also emerging as a constructive and potentially beneficial therapeutic strategy. We will discuss the role of proteoglycans in selected and emerging areas of proteoglycan science, such as neurodegenerative diseases, autophagy, angiogenesis, cancer, infections and their impact on mammalian diseases.
Collapse
Affiliation(s)
- Christopher Xie
- Department of Pathology and Genomic Medicine, the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
7
|
Chen Y, Dufour CR, Han L, Li T, Xia H, Giguère V. Hierarchical Phosphorylation of HOXB13 by mTOR Dictates Its Activity and Oncogenic Function in Prostate Cancer. Mol Cancer Res 2023; 21:1050-1063. [PMID: 37409967 PMCID: PMC10544006 DOI: 10.1158/1541-7786.mcr-23-0086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/23/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Dysregulation of mTOR signaling plays a critical role in promoting prostate cancer growth. HOXB13, a homeodomain transcription factor, is known to influence the androgen response and prostate cancer development. Recently, HOXB13 was found to complex with mTOR on chromatin. However, the functional crosstalk between HOXB13 and mTOR remains elusive. We now report that mTOR directly interacts with and hierarchically phosphorylates HOXB13 at threonine 8 and 41 then serine 31 to promote its interaction with the E3 ligase SKP2 while enhancing its oncogenic properties. Expression of HOXB13 harboring phosphomimetic mutations at the mTOR-targeted sites stimulates prostate cancer cellular growth both in vitro and in murine xenografts. Transcriptional profiling studies revealed a phospho-HOXB13-dependent gene signature capable of robustly discriminating between normal prostate tissues, primary and metastatic prostate cancer samples. This work uncovers a previously unanticipated molecular cascade by which mTOR directly phosphorylates HOXB13 to dictate a specific gene program with oncogenic implications in prostate cancer. IMPLICATIONS Control of HOXB13 transcriptional activity via its direct phosphorylation by the mTOR kinase is a potential therapeutic avenue for the management of advanced prostate cancer.
Collapse
Affiliation(s)
- Yonghong Chen
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | | | - Lingwei Han
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Ting Li
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
| | - Hui Xia
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Vincent Giguère
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| |
Collapse
|
8
|
Mondal DK, Xie C, Buraschi S, Iozzo RV. Decorin suppresses tumor lymphangiogenesis: A mechanism to curtail cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555187. [PMID: 37693608 PMCID: PMC10491239 DOI: 10.1101/2023.08.28.555187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The complex interplay between malignant cells and the cellular and molecular components of the tumor stroma is a key aspect of cancer growth and development. These tumor-host interactions are often affected by soluble bioactive molecules such as proteoglycans. Decorin, an archetypical small leucine-rich proteoglycan primarily expressed by stromal cells, affects cancer growth in its soluble form by interacting with several receptor tyrosine kinases (RTK). Overall, decorin leads to a context-dependent and protracted cessation of oncogenic RTK activity by attenuating their ability to drive a pro-survival program and to sustain a pro-angiogenic network. Through an unbiased transcriptomic analysis using deep RNAseq, we discovered that decorin downregulated a cluster of tumor-associated genes involved in lymphatic vessel development when systemically delivered to mice harboring breast carcinoma allografts. We found that Lyve1 and Podoplanin, two established markers of lymphatic vessels, were markedly suppressed at both the mRNA and protein levels and this suppression correlated with a significant reduction in tumor lymphatic vessels. We further discovered that soluble decorin, but not its homologous proteoglycan biglycan, inhibited lymphatic vessel sprouting in an ex vivo 3D model of lymphangiogenesis. Mechanistically, we found that decorin interacted with VEGFR3, the main lymphatic RTK, and its activity was required for the decorin-mediated block of lymphangiogenesis. Finally, we discovered that Lyve1 was in part degraded via decorin-evoked autophagy in a nutrient- and energy-independent manner. These findings implicate decorin as a new biological factor with anti-lymphangiogenic activity and provide a potential therapeutic agent for curtailing breast cancer growth and metastasis.
Collapse
|
9
|
Surendran H, Palaniyandi T, Natarajan S, Hari R, Viwanathan S, Baskar G, Abdul Wahab MR, Ravi M, Rajendran BK. Role of homeobox d10 gene targeted signaling pathways in cancers. Pathol Res Pract 2023; 248:154643. [PMID: 37406379 DOI: 10.1016/j.prp.2023.154643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
Homeobox D10 (HOXD10) is a transcription factor from the homeobox gene family that controls cell differentiation and morphogenesis throughout development.Due to their functional interaction, changes in HOXD10 gene expression might induce tumors. This narrative review focuses on how and why the dysregulation in the signaling pathways linked with HOXD10 contributes to the metastatic development of cancer. Organ development and tissue homeostasis need highly conserved homeotic transcription factors from homeobox (HOX) genes. Their dysregulation disrupts regulatory molecule action, causing tumors. The HOXD10 gene is upregulated in breast, gastric, hepatocellular, colorectal, bladder, cholangiocellular carcinoma and prostate cancer. Tumor signaling pathways are affected by HOXD10 gene expression changes. This study examines HOXD10-associated signaling pathway dysregulation, which may alter metastatic cancer signaling. In addition, the theoretical foundations that alter HOXD10-mediated therapeutic resistance in malignancies has been presented. New cancer therapy methods will be simpler to develop with the newly discovered knowledge. This review showed that HOXD10 may be a tumor suppressor gene and a new cancer treatment target signaling pathway.
Collapse
Affiliation(s)
- Hemapreethi Surendran
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India; Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, Tamilnadu, India.
| | - Sudhakar Natarajan
- Department of Virology and Biotechnology, ICMR - National institute for Research in Tuberculosis (NIRT), Chetpet, Chennai 600031 Tamil Nadu, India
| | - Rajeswary Hari
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Sandhiya Viwanathan
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Mugip Rahaman Abdul Wahab
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116 Tamil Nadu, India
| | | |
Collapse
|
10
|
A Systematic Pan-Cancer Analysis of MEIS1 in Human Tumors as Prognostic Biomarker and Immunotherapy Target. J Clin Med 2023; 12:jcm12041646. [PMID: 36836180 PMCID: PMC9964192 DOI: 10.3390/jcm12041646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND We intended to explore the potential immunological functions and prognostic value of Myeloid Ecotropic Viral Integration Site 1 (MEIS1) across 33 cancer types. METHODS The data were acquired from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx) and Gene expression omnibus (GEO) datasets. Bioinformatics was used to excavate the potential mechanisms of MEIS1 across different cancers. RESULTS MEIS1 was downregulated in most tumors, and it was linked to the immune infiltration level of cancer patients. MEIS1 expression was different in various immune subtypes including C2 (IFN-gamma dominant), C5 (immunologically quiet), C3 (inflammatory), C4 (lymphocyte depleted), C6 (TGF-b dominant) and C1 (wound healing) in various cancers. MEIS1 expression was correlated with Macrophages_M2, CD8+T cells, Macrophages_M1, Macrophages_M0 and neutrophils in many cancers. MEIS1 expression was negatively related to tumor mutational burden (TMB), microsatellite instability (MSI) and neoantigen (NEO) in several cancers. Low MEIS1 expression predicts poor overall survival (OS) in adrenocortical carcinoma (ACC), head and neck squamous cell carcinoma (HNSC), and kidney renal clear cell carcinoma (KIRC) patients, while high MEIS1 expression predicts poor OS in colon adenocarcinoma (COAD) and low grade glioma (LGG) patients. CONCLUSION Our findings revealed that MEIS1 is likely to be a potential new target for immuno-oncology.
Collapse
|
11
|
Wang S, Sun Y, Liu X, Guo Y, Huang Y, Zhang S, Tian Q. Meis1 Controls the Differentiation of Eye Progenitor Cells and the Formation of Posterior Poles during Planarian Regeneration. Int J Mol Sci 2023; 24:ijms24043505. [PMID: 36834910 PMCID: PMC9961902 DOI: 10.3390/ijms24043505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/21/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
As a member of TALE family, Meis1 has been proven to regulate cell proliferation and differentiation during cell fate commitment; however, the mechanism is still not fully understood. The planarian, which has an abundance of stem cells (neoblasts) responsible for regenerating any organ after injury, is an ideal model for studying the mechanisms of tissue identity determination. Here, we characterized a planarian homolog of Meis1 from the planarian Dugesia japonica. Importantly, we found that knockdown of DjMeis1 inhibits the differentiation of neoblasts into eye progenitor cells and results in an eyeless phenotype with normal central nervous system. Furthermore, we observed that DjMeis1 is required for the activation of Wnt signaling pathway by promoting the Djwnt1 expression during posterior regeneration. The silencing of DjMeis1 suppresses the expression of Djwnt1 and results in the inability to reconstruct posterior poles. In general, our findings indicated that DjMeis1 acts as a trigger for the activation of eye and tail regeneration by regulating the differentiation of eye progenitor cells and the formation of posterior poles, respectively.
Collapse
Affiliation(s)
- Shaocong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yujia Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomai Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yajun Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongding Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shoutao Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou 450046, China
- Correspondence: (S.Z.); (Q.T.)
| | - Qingnan Tian
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (S.Z.); (Q.T.)
| |
Collapse
|
12
|
Otálora-Otálora BA, López-Kleine L, Rojas A. Lung Cancer Gene Regulatory Network of Transcription Factors Related to the Hallmarks of Cancer. Curr Issues Mol Biol 2023; 45:434-464. [PMID: 36661515 PMCID: PMC9857713 DOI: 10.3390/cimb45010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
The transcriptomic analysis of microarray and RNA-Seq datasets followed our own bioinformatic pipeline to identify a transcriptional regulatory network of lung cancer. Twenty-six transcription factors are dysregulated and co-expressed in most of the lung cancer and pulmonary arterial hypertension datasets, which makes them the most frequently dysregulated transcription factors. Co-expression, gene regulatory, coregulatory, and transcriptional regulatory networks, along with fibration symmetries, were constructed to identify common connection patterns, alignments, main regulators, and target genes in order to analyze transcription factor complex formation, as well as its synchronized co-expression patterns in every type of lung cancer. The regulatory function of the most frequently dysregulated transcription factors over lung cancer deregulated genes was validated with ChEA3 enrichment analysis. A Kaplan-Meier plotter analysis linked the dysregulation of the top transcription factors with lung cancer patients' survival. Our results indicate that lung cancer has unique and common deregulated genes and transcription factors with pulmonary arterial hypertension, co-expressed and regulated in a coordinated and cooperative manner by the transcriptional regulatory network that might be associated with critical biological processes and signaling pathways related to the acquisition of the hallmarks of cancer, making them potentially relevant tumor biomarkers for lung cancer early diagnosis and targets for the development of personalized therapies against lung cancer.
Collapse
Affiliation(s)
- Beatriz Andrea Otálora-Otálora
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia
- Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 11001, Colombia
| | - Liliana López-Kleine
- Departamento de Estadística, Universidad Nacional de Colombia, Bogotá 11001, Colombia
- Correspondence: (L.L.-K.); (A.R.)
| | - Adriana Rojas
- Facultad de Medicina, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá 110211, Colombia
- Correspondence: (L.L.-K.); (A.R.)
| |
Collapse
|
13
|
Jia Y, Reboulet J, Gillet B, Hughes S, Forcet C, Tribollet V, Hajj Sleiman N, Kundlacz C, Vanacker JM, Bleicher F, Merabet S. A Live Cell Protein Complementation Assay for ORFeome-Wide Probing of Human HOX Interactomes. Cells 2023; 12:cells12010200. [PMID: 36611993 PMCID: PMC9818449 DOI: 10.3390/cells12010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
Biological pathways rely on the formation of intricate protein interaction networks called interactomes. Getting a comprehensive map of interactomes implies the development of tools that allow one to capture transient and low-affinity protein-protein interactions (PPIs) in live conditions. Here we presented an experimental strategy: the Cell-PCA (cell-based protein complementation assay), which was based on bimolecular fluorescence complementation (BiFC) for ORFeome-wide screening of proteins that interact with different bait proteins in the same live cell context, by combining high-throughput sequencing method. The specificity and sensitivity of the Cell-PCA was established by using a wild-type and a single-amino-acid-mutated HOXA9 protein, and the approach was subsequently applied to seven additional human HOX proteins. These proof-of-concept experiments revealed novel molecular properties of HOX interactomes and led to the identification of a novel cofactor of HOXB13 that promoted its proliferative activity in a cancer cell context. Taken together, our work demonstrated that the Cell-PCA was pertinent for revealing and, importantly, comparing the interactomes of different or highly related bait proteins in the same cell context.
Collapse
Affiliation(s)
- Yunlong Jia
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Jonathan Reboulet
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
- LiPiCs, 46 Allée d’Italie, 69007 Lyon, France
| | - Benjamin Gillet
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Sandrine Hughes
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Christelle Forcet
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Violaine Tribollet
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Nawal Hajj Sleiman
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Cindy Kundlacz
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Jean-Marc Vanacker
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
| | - Françoise Bleicher
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
- Correspondence: franç (F.B.); (S.M.)
| | - Samir Merabet
- IGFL, CNRS UMR5242, ENS-Lyon, UCBL-1, INRA USC1370, 32 Av. Tony Garnier, 69007 Lyon, France
- Correspondence: franç (F.B.); (S.M.)
| |
Collapse
|
14
|
Trendowski MR, Sample C, Baird T, Sadeghpour A, Moon D, Ruterbusch JJ, Beebe-Dimmer JL, Cooney KA. Germline Variants in DNA Damage Repair Genes and HOXB13 Among Black Patients With Early-Onset Prostate Cancer. JCO Precis Oncol 2022; 6:e2200460. [PMID: 36446039 PMCID: PMC9812633 DOI: 10.1200/po.22.00460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Genetic studies of prostate cancer susceptibility have predominantly focused on non-Hispanic White men, despite the observation that Black men are more likely to develop prostate cancer and die from the disease. Therefore, we sought to identify genetic variants in Black patients diagnosed with early-onset prostate cancer. METHODS Whole-exome sequencing of germline DNA from a population-based cohort of Black men diagnosed with prostate cancer at age 62 years or younger was performed. Analysis was focused on a panel of DNA damage repair (DDR) genes and HOXB13. All discovered variants were ranked according to their pathogenic potential based upon REVEL score, evidence from existing literature, and prevalence in the cohort. Logistic regression was used to investigate associations between mutation status and relevant clinical characteristics. RESULTS Among 743 Black prostate cancer patients, we identified 26 unique pathogenic (P) or likely pathogenic (LP) variants in 14 genes (including HOXB13, BRCA1/2, BRIP1, ATM, CHEK2, and PALB2) among 30 men, or approximately 4.0% of the patient population. We also identified 33 unique variants of unknown significance in 16 genes among 39 men. Because of the rarity of these variants in the population, most associations between clinical characteristics did not achieve statistical significance. However, our results suggest that carriers for P or LP (P/LP) variants were more likely to have a first-degree relative diagnosed with DDR gene-associated cancer, have a higher prostate-specific antigen at time of diagnosis, and be diagnosed with metastatic disease. CONCLUSION Variants in DDR genes and HOXB13 may be important cancer risk factors for Black men diagnosed with early-onset prostate cancer, and are more frequently observed in men with a family history of cancer.
Collapse
Affiliation(s)
| | | | - Tara Baird
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Azita Sadeghpour
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - David Moon
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Julie J. Ruterbusch
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Jennifer L. Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Kathleen A. Cooney
- Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke Cancer Institute, Durham, NC
| |
Collapse
|
15
|
Pfeifer B, Saranti A, Holzinger A. GNN-SubNet: disease subnetwork detection with explainable graph neural networks. Bioinformatics 2022; 38:ii120-ii126. [PMID: 36124793 DOI: 10.1093/bioinformatics/btac478] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MOTIVATION The tremendous success of graphical neural networks (GNNs) already had a major impact on systems biology research. For example, GNNs are currently being used for drug target recognition in protein-drug interaction networks, as well as for cancer gene discovery and more. Important aspects whose practical relevance is often underestimated are comprehensibility, interpretability and explainability. RESULTS In this work, we present a novel graph-based deep learning framework for disease subnetwork detection via explainable GNNs. Each patient is represented by the topology of a protein-protein interaction (PPI) network, and the nodes are enriched with multi-omics features from gene expression and DNA methylation. In addition, we propose a modification of the GNNexplainer that provides model-wide explanations for improved disease subnetwork detection. AVAILABILITY AND IMPLEMENTATION The proposed methods and tools are implemented in the GNN-SubNet Python package, which we have made available on our GitHub for the international research community (https://github.com/pievos101/GNN-SubNet). SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Bastian Pfeifer
- Institute for Medical Informatics Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Anna Saranti
- Institute for Medical Informatics Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Andreas Holzinger
- Institute for Medical Informatics Statistics and Documentation, Medical University Graz, Graz, Austria.,Human-Centered AI Lab, Department of Forest- and Soil Sciences, University of Natural Resources and Life Sciences Vienna, Vienna, Austria.,Alberta Machine Intelligence Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
Lagler DK, Hannemann E, Eck K, Klawatsch J, Seichter D, Russ I, Mendel C, Lühken G, Krebs S, Blum H, Upadhyay M, Medugorac I. Fine-mapping and identification of candidate causal genes for tail length in the Merinolandschaf breed. Commun Biol 2022; 5:918. [PMID: 36068271 PMCID: PMC9448734 DOI: 10.1038/s42003-022-03854-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022] Open
Abstract
Docking the tails of lambs in long-tailed sheep breeds is a common practice worldwide. But this practice is associated with pain. Breeding for a shorter tail could offer an alternative. Therefore, this study aimed to analyze the natural tail length variation in the Merinolandschaf and to identify causal alleles for the short tail phenotype segregating within long-tailed breeds. We used SNP-based association analysis and haplotype-based mapping in 362 genotyped (Illumina OvineSNP50) and phenotyped Merinolandschaf lambs. Genome-wide significant regions were capture sequenced in 48 lambs and comparatively analyzed in various long and short-tailed sheep breeds and wild sheep subspecies. Here we show a SNP located in the first exon of HOXB13 and a SINE element located in the promotor of HOXB13 as promising candidates. These results enable more precise breeding towards shorter tails, improve animal welfare by amplification of ancestral alleles and contribute to a better understanding of differential embryonic development.
Collapse
Affiliation(s)
- Dominik Karl Lagler
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany
- Tierzuchtforschung e.V. München, Senator-Gerauer-Str. 23, 85586, Poing, Germany
| | - Elisabeth Hannemann
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany
| | - Kim Eck
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany
- Tierzuchtforschung e.V. München, Senator-Gerauer-Str. 23, 85586, Poing, Germany
| | - Jürgen Klawatsch
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany
- Tierzuchtforschung e.V. München, Senator-Gerauer-Str. 23, 85586, Poing, Germany
| | - Doris Seichter
- Tierzuchtforschung e.V. München, Senator-Gerauer-Str. 23, 85586, Poing, Germany
| | - Ingolf Russ
- Tierzuchtforschung e.V. München, Senator-Gerauer-Str. 23, 85586, Poing, Germany
| | - Christian Mendel
- Institute for Animal Breeding, Bavarian State Research Center for Agriculture, Prof.-Dürrwaechter-Platz 1, 85586, Poing, Germany
| | - Gesine Lühken
- Institute of Animal Breeding and Genetics, JLU Gießen, Ludwigstr. 21, 35390, Gießen, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-University Munich, 80539, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-University Munich, 80539, Munich, Germany
| | - Maulik Upadhyay
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany
| | - Ivica Medugorac
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Lena-Christ-Str. 48, 82152, Martinsried, Germany.
| |
Collapse
|
17
|
Setti Boubaker N, Gurtner A, Trabelsi N, Manni I, Blel A, Saadi A, Chakroun M, Naimi Z, Zaghbib S, Ksontini M, Meddeb K, Rammeh S, Ayed H, Chebil M, Piaggio G, Ouerhani S. An insight into the diagnostic and prognostic value of
HOX A13
’s expression in non‐muscle invasive bladder cancer. J Clin Lab Anal 2022; 36:e24606. [PMID: 35853090 PMCID: PMC9459288 DOI: 10.1002/jcla.24606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/15/2022] Open
Abstract
Background Several studies have interrogated the molecular pathways and their interacting genes underlying bladder cancer (BCa) tumorigenesis, yet, the role of homeobox genes is still poorly understood. Specifically, HOXA13, which plays an important role as a major actor in the urogenital tract's development. Methods Immunohistochemical (IHC) staining was performed to inspect the differential expression of HOXA13 protein in non‐muscle‐invasive bladder cancer (NMIBC) and non‐tumoral tissues. A semiquantitative scoring system was adopted to evaluate the IHC labeling. Correlation to clinical parameters was performed by descriptive statistics. Overall survival was estimated by the Kaplan–Meier method and Cox regression model. The functional HOX A13 protein association networks (PPI) were obtained using String 11.0 database. Results HOX A13 exhibited cytoplasmic and nuclear staining. Its expression levels were lower in high‐grade NMIBC (HG NMIBC) compared to low‐grade ones (LG NMIBC). The expression of HOX A13 was correlated to tumor grade (LG/HG) (p = 0.036) and stage (TA/T1) (p = 0.036). Nevertheless, its expression was not correlated to clinical parameters and was not able to predict the overall survival of patients with HG NMIBC. Finally, PPI analysis revealed that HOX A13 seems to be a part of a molecular network holding mainly PBX1, MEIS, ALDH1A2, HOX A10, and HOX A11. Conclusion The deregulation of HOX A13 is not associated with the prognosis of BCa. It seems to be rather implicated in the early initiation of urothelial tumorigenesis and thus may serve as a diagnostic marker in patients with NMIBC. Further experimentations on larger validation sets are mandatory.
Collapse
Affiliation(s)
- Nouha Setti Boubaker
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP‐MB) INSAT University of Tunis Carthage Tunis Tunisia
- UOSD SAFU Department of Research, Diagnosis and Innovative Technologies IRCCS‐Regina Elena National Cancer Institute Rome Italy
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Aymone Gurtner
- UOSD SAFU Department of Research, Diagnosis and Innovative Technologies IRCCS‐Regina Elena National Cancer Institute Rome Italy
- Institute of Translational Pharmacology (IFT) National Research Council (CNR) Rome Italy
| | - Nesrine Trabelsi
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP‐MB) INSAT University of Tunis Carthage Tunis Tunisia
| | - Isabella Manni
- UOSD SAFU Department of Research, Diagnosis and Innovative Technologies IRCCS‐Regina Elena National Cancer Institute Rome Italy
| | - Ahlem Blel
- Pathology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis El Manar Tunis Tunisia
| | - Ahmed Saadi
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Marouene Chakroun
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Zeineb Naimi
- Medical Oncology Department Faculty of Medicine Salah Azaiez Institute University of Tunis‐El Manar Tunis Tunisia
| | - Selim Zaghbib
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Meriam Ksontini
- Pathology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis El Manar Tunis Tunisia
| | - Khedija Meddeb
- Medical Oncology Department Faculty of Medicine Salah Azaiez Institute University of Tunis‐El Manar Tunis Tunisia
| | - Soumaya Rammeh
- Pathology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis El Manar Tunis Tunisia
| | - Haroun Ayed
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Mohamed Chebil
- Urology Department Faculty of Medicine Charles Nicolle Hospital University of Tunis‐El Manar Tunis Tunisia
| | - Giulia Piaggio
- UOSD SAFU Department of Research, Diagnosis and Innovative Technologies IRCCS‐Regina Elena National Cancer Institute Rome Italy
| | - Slah Ouerhani
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP‐MB) INSAT University of Tunis Carthage Tunis Tunisia
| |
Collapse
|
18
|
Zhong ME, Huang ZP, Wang X, Cai D, Li CH, Gao F, Wu XJ, Wang W. A Transcription Factor Signature Can Identify the CMS4 Subtype and Stratify the Prognostic Risk of Colorectal Cancer. Front Oncol 2022; 12:902974. [PMID: 35847938 PMCID: PMC9280271 DOI: 10.3389/fonc.2022.902974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundColorectal cancer (CRC) is a heterogeneous disease, and current classification systems are insufficient for stratifying patients with different risks. This study aims to develop a generalized, individualized prognostic consensus molecular subtype (CMS)-transcription factors (TFs)-based signature that can predict the prognosis of CRC.MethodsWe obtained differentially expressed TF signature and target genes between the CMS4 and other CMS subtypes of CRC from The Cancer Genome Atlas (TCGA) database. A multi-dimensional network inference integrative analysis was conducted to identify the master genes and establish a CMS4-TFs-based signature. For validation, an in-house clinical cohort (n = 351) and another independent public CRC cohort (n = 565) were applied. Gene set enrichment analysis (GSEA) and prediction of immune cell infiltration were performed to interpret the biological significance of the model.ResultsA CMS4-TFs-based signature termed TF-9 that includes nine TF master genes was developed. Patients in the TF-9 high-risk group have significantly worse survival, regardless of clinical characteristics. The TF-9 achieved the highest mean C-index (0.65) compared to all other signatures reported (0.51 to 0.57). Immune infiltration revealed that the microenvironment in the high-risk group was highly immune suppressed, as evidenced by the overexpression of TIM3, CD39, and CD40, suggesting that high-risk patients may not directly benefit from the immune checkpoint inhibitors.ConclusionsThe TF-9 signature allows a more precise categorization of patients with relevant clinical and biological implications, which may be a valuable tool for improving the tailoring of therapeutic interventions in CRC patients.
Collapse
Affiliation(s)
- Min-Er Zhong
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Ping Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xun Wang
- Department of Biomedical Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng-Hang Li
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wei Wang, ; Xiao-Jian Wu, ; Feng Gao,
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wei Wang, ; Xiao-Jian Wu, ; Feng Gao,
| | - Wei Wang
- Biomedical Big Data Centre, Department of Gynaecology, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
- *Correspondence: Wei Wang, ; Xiao-Jian Wu, ; Feng Gao,
| |
Collapse
|
19
|
Lu X, Fong KW, Gritsina G, Wang F, Baca SC, Brea LT, Berchuck JE, Spisak S, Ross J, Morrissey C, Corey E, Chandel NS, Catalona WJ, Yang X, Freedman ML, Zhao JC, Yu J. HOXB13 suppresses de novo lipogenesis through HDAC3-mediated epigenetic reprogramming in prostate cancer. Nat Genet 2022; 54:670-683. [PMID: 35468964 PMCID: PMC9117466 DOI: 10.1038/s41588-022-01045-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/28/2022] [Indexed: 01/16/2023]
Abstract
HOXB13, a homeodomain transcription factor, critically regulates androgen receptor (AR) activities and androgen-dependent prostate cancer (PCa) growth. However, its functions in AR-independent contexts remain elusive. Here we report HOXB13 interaction with histone deacetylase HDAC3, which is disrupted by the HOXB13 G84E mutation that has been associated with early-onset PCa. Independently of AR, HOXB13 recruits HDAC3 to lipogenic enhancers to catalyze histone deacetylation and suppress lipogenic regulators such as fatty acid synthase. Analysis of human tissues reveals that the HOXB13 gene is hypermethylated and downregulated in approximately 30% of metastatic castration-resistant PCa. HOXB13 loss or G84E mutation leads to lipid accumulation in PCa cells, thereby promoting cell motility and xenograft tumor metastasis, which is mitigated by pharmaceutical inhibition of fatty acid synthase. In summary, we present evidence that HOXB13 recruits HDAC3 to suppress de novo lipogenesis and inhibit tumor metastasis and that lipogenic pathway inhibitors may be useful to treat HOXB13-low PCa.
Collapse
Affiliation(s)
- Xiaodong Lu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ka-wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Galina Gritsina
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Fang Wang
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sylvan C. Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lourdes T. Brea
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jacob E. Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sandor Spisak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jenny Ross
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, USA
| | - Navdeep S. Chandel
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA,Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University, Chicago, IL, USA,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - William J. Catalona
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA,Department of Urology, Northwestern University, Chicago, IL, USA
| | - Ximing Yang
- Department of Pathology, Northwestern University, Chicago, IL, USA,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan C. Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Co-Corresponding Authors: Jindan Yu, M.D., Ph.D. , Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine; Jonathan C. Zhao,
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA,Co-Corresponding Authors: Jindan Yu, M.D., Ph.D. , Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine; Jonathan C. Zhao,
| |
Collapse
|
20
|
Xie C, Mondal DK, Ulas M, Neill T, Iozzo RV. Oncosuppressive roles of decorin through regulation of multiple receptors and diverse signaling pathways. Am J Physiol Cell Physiol 2022; 322:C554-C566. [PMID: 35171698 PMCID: PMC8917911 DOI: 10.1152/ajpcell.00016.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Decorin is a stromal-derived prototype member of the small leucine-rich proteoglycan gene family. In addition to its functions as a regulator of collagen fibrillogenesis and TGF-β activity soluble decorin acts as a pan-receptor tyrosine kinase (RTK) inhibitor. Decorin binds to various RTKs including EGFR HER2 HGFR/Met VEGFR2 TLR and IGFR. Although the molecular mechanism for the action of decorin on these receptors is not entirely elucidated overall decorin evokes transient activation of these receptors with suppression of downstream signaling cascades culminating in growth inhibition followed by their physical downregulation via caveosomal internalization and degradation. In the case of Met decorin leads to decreased β-catenin signaling pathway and growth suppression. As most of these RTKs are responsible for providing a growth advantage to cancer cells the result of decorin treatment is oncosuppression. Another decorin-driven mechanism to restrict cancer growth and dissemination is by impeding angiogenesis via vascular endothelial growth factor receptor 2 (VEGFR2) and the concurrent activation of protracted endothelial cell autophagy. In this review we will dissect the multiple roles of decorin in cancer biology and its potential use as a next-generation protein-based adjuvant therapy to combat cancer.
Collapse
Affiliation(s)
- Christopher Xie
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dipon K. Mondal
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mikdat Ulas
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Morgan R, Hunter K, Pandha HS. Downstream of the HOX genes: explaining conflicting tumour suppressor and oncogenic functions in cancer. Int J Cancer 2022; 150:1919-1932. [PMID: 35080776 PMCID: PMC9304284 DOI: 10.1002/ijc.33949] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 01/07/2022] [Indexed: 11/07/2022]
Abstract
The HOX genes are a highly conserved group of transcription factors that have key roles in early development, but which are also highly expressed in most cancers. Many studies have found strong associative relationships between the expression of individual HOX genes in tumours and clinical parameters including survival. For the majority of HOX genes, high tumour expression levels seem to be associated with a worse outcome for patients, and in some cases this has been shown to result from the activation of pro-oncogenic genes and pathways. However, there are also many studies that indicate a tumour suppressor role for some HOX genes, sometimes with conclusions that contradict earlier work. In this review, we have attempted to clarify the role of HOX genes in cancer by focusing on their downstream targets as identified in studies that provide experimental evidence for their activation or repression. On this basis, the majority of HOX genes would appear to have a pro-oncogenic function, with the notable exception of HOXD10, which acts exclusively as a tumour suppressor. HOX proteins regulate a wide range of target genes involved in metastasis, cell death, proliferation, and angiogenesis, and activate key cell signalling pathways. Furthermore, for some functionally related targets, this regulation is achieved by a relatively small subgroup of HOX genes.
Collapse
Affiliation(s)
- Richard Morgan
- School of Biomedical SciencesUniversity of West LondonLondonUK
| | - Keith Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical DentistryUniversity of SheffieldSheffieldUK
| | - Hardev S. Pandha
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| |
Collapse
|
22
|
Feng Y, Zhang T, Wang Y, Xie M, Ji X, Luo X, Huang W, Xia L. Homeobox Genes in Cancers: From Carcinogenesis to Recent Therapeutic Intervention. Front Oncol 2021; 11:770428. [PMID: 34722321 PMCID: PMC8551923 DOI: 10.3389/fonc.2021.770428] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
The homeobox (HOX) genes encoding an evolutionarily highly conserved family of homeodomain-containing transcriptional factors are essential for embryogenesis and tumorigenesis. HOX genes are involved in cell identity determination during early embryonic development and postnatal processes. The deregulation of HOX genes is closely associated with numerous human malignancies, highlighting the indispensable involvement in mortal cancer development. Since most HOX genes behave as oncogenes or tumor suppressors in human cancer, a better comprehension of their upstream regulators and downstream targets contributes to elucidating the function of HOX genes in cancer development. In addition, targeting HOX genes may imply therapeutic potential. Recently, novel therapies such as monoclonal antibodies targeting tyrosine receptor kinases, small molecular chemical inhibitors, and small interfering RNA strategies, are difficult to implement for targeting transcriptional factors on account of the dual function and pleiotropic nature of HOX genes-related molecular networks. This paper summarizes the current state of knowledge on the roles of HOX genes in human cancer and emphasizes the emerging importance of HOX genes as potential therapeutic targets to overcome the limitations of present cancer therapy.
Collapse
Affiliation(s)
- Yangyang Feng
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongyue Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Xie
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Ji
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Blasi F, Bruckmann C. MEIS1 in Hematopoiesis and Cancer. How MEIS1-PBX Interaction Can Be Used in Therapy. J Dev Biol 2021; 9:jdb9040044. [PMID: 34698191 PMCID: PMC8544432 DOI: 10.3390/jdb9040044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 11/26/2022] Open
Abstract
Recently MEIS1 emerged as a major determinant of the MLL-r leukemic phenotype. The latest and most efficient drugs effectively decrease the levels of MEIS1 in cancer cells. Together with an overview of the latest drugs developed to target MEIS1 in MLL-r leukemia, we review, in detail, the role of MEIS1 in embryonic and adult hematopoiesis and suggest how a more profound knowledge of MEIS1 biochemistry can be used to design potent and effective drugs against MLL-r leukemia. In addition, we present data showing that the interaction between MEIS1 and PBX1 can be blocked efficiently and might represent a new avenue in anti-MLL-r and anti-leukemic therapy.
Collapse
|
24
|
Geng H, Liu G, Hu J, Li J, Wang D, Zou S, Xu X. HOXB13 suppresses proliferation, migration and invasion, and promotes apoptosis of gastric cancer cells through transcriptional activation of VGLL4 to inhibit the involvement of TEAD4 in the Hippo signaling pathway. Mol Med Rep 2021; 24:722. [PMID: 34396425 PMCID: PMC8383034 DOI: 10.3892/mmr.2021.12361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer (GC) is one of the most common types of malignancy worldwide and is accompanied by both high mortality and morbidity rates. Homeobox B13 (HOXB13) has been reported to act as a tumor suppressor gene in multiple types of human cancer. The present study aimed to investigate the effects and potential underlying molecular mechanisms of HOXB13 in the progression of GC. The expression of HOXB13 in GC cells was first examined using the Cancer Cell Line Encyclopedia database and subsequently validated in a number of GC cell lines. Following HOXB13 overexpression (Ov-HOXB13), HGC-27 cell proliferation was evaluated by colony formation and Cell Counting Kit-8 assays. Wound healing and Matrigel assays were used to determine the migratory and invasive abilities, respectively. Additionally, cell apoptosis was assessed using TUNEL staining, and the expression of apoptosis-related proteins was detected by western blot analysis. Subsequently, TEA domain transcription factor 4 (TEAD4) was overexpressed to evaluate the effects on HGC-27 cell proliferation, migration, invasion and apoptosis following co-transfection with Ov-HOXB13. The potential binding sites of HOXB13 on the vestigial-like family member 4 (VGLL4) promoter were verified using chromatin immunoprecipitation and dual luciferase reporter assays. Moreover, the expression levels of proteins involved in the Hippo signaling pathway were analyzed using western blotting. The results revealed that the expression of HOXB13 was notably lower in GC cells compared with normal gastric cells. The overexpression of HOXB13 significantly inhibited the proliferation, migration and invasion, but promoted the apoptosis of HGC-27 cells. Moreover, Ov-HOXB13 downregulated TEAD4 expression. Notably, Ov-TEAD4 transfection partially reversed the effects of Ov-HOXB13 on the cellular behaviors of HGC-27 cells. HOXB13 was also confirmed to bind with the VGLL4 promoter. The knockdown of VGLL4 restored the inhibitory effects of Ov-HOXB13 on the expression levels of VGLL4 and Hippo pathway signaling proteins. In conclusion, the findings of the present study suggested that Ov-HOXB13 may suppress the proliferation, migration and invasion, and promote the apoptosis of GC cells through the transcriptional activation of VGLL4 to inhibit the involvement of TEAD4 in the Hippo signaling pathway. These results may provide novel and potent targets for the treatment of GC.
Collapse
Affiliation(s)
- Hui Geng
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Guangli Liu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Jiandong Hu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Jianchao Li
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Dong Wang
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Shenshan Zou
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| | - Xintao Xu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
25
|
Yao M, Gu Y, Yang Z, Zhong K, Chen Z. MEIS1 and its potential as a cancer therapeutic target (Review). Int J Mol Med 2021; 48:181. [PMID: 34318904 PMCID: PMC8354308 DOI: 10.3892/ijmm.2021.5014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/08/2021] [Indexed: 01/26/2023] Open
Abstract
Meis homeobox 1 (Meis1) was initially discovered in 1995 as a factor involved in leukemia in an animal model. Subsequently, 2 years later, MEIS1, the human homolog, was cloned in the liver and cerebellum, and was found to be highly expressed in myeloid leukemia cells. The MEIS1 gene, located on chromosome 2p14, encodes a 390-amino acid protein with six domains. The expression of homeobox protein MEIS1 is affected by cell type, age and environmental conditions, as well as the pathological state. Certain types of modifications of MEIS1 and its protein interaction with homeobox or pre-B-cell leukemia homeobox proteins have been described. As a transcription factor, MEIS1 protein is involved in cell proliferation in leukemia and some solid tumors. The present review article discusses the molecular biology, modifications, protein-protein interactions, as well as the role of MEIS1 in cell proliferation of cancer cells and MEIS1 inhibitors. It is suggested by the available literature MEIS1 has potential to become a cancer therapeutic target.
Collapse
Affiliation(s)
- Maozhong Yao
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Zhaoxin Yang
- Teaching Experimental Animal Center, Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Keyan Zhong
- Teaching Experimental Animal Center, Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Zhanjuan Chen
- Chemical Experiment Teaching Center, College of Pharmacy, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| |
Collapse
|
26
|
Neill T, Kapoor A, Xie C, Buraschi S, Iozzo RV. A functional outside-in signaling network of proteoglycans and matrix molecules regulating autophagy. Matrix Biol 2021; 100-101:118-149. [PMID: 33838253 PMCID: PMC8355044 DOI: 10.1016/j.matbio.2021.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Proteoglycans and selected extracellular matrix constituents are emerging as intrinsic and critical regulators of evolutionarily conversed, intracellular catabolic pathways. Often, these secreted molecules evoke sustained autophagy in a variety of cell types, tissues, and model systems. The unique properties of proteoglycans have ushered in a paradigmatic shift to broaden our understanding of matrix-mediated signaling cascades. The dynamic cellular pathway controlling autophagy is now linked to an equally dynamic and fluid signaling network embedded in a complex meshwork of matrix molecules. A rapidly emerging field of research encompasses multiple matrix-derived candidates, representing a menagerie of soluble matrix constituents including decorin, biglycan, endorepellin, endostatin, collagen VI and plasminogen kringle 5. These matrix constituents are pro-autophagic and simultaneously anti-angiogenic. In contrast, perlecan, laminin α2 chain, and lumican have anti-autophagic functions. Mechanistically, each matrix constituent linked to intracellular catabolic events engages a specific cell surface receptor that often converges on a common core of the autophagic machinery including AMPK, Peg3 and Beclin 1. We consider this matrix-evoked autophagy as non-canonical given that it occurs in an allosteric manner and is independent of nutrient availability or prevailing bioenergetics control. We propose that matrix-regulated autophagy is an important outside-in signaling mechanism for proper tissue homeostasis that could be therapeutically leveraged to combat a variety of diseases.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christopher Xie
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
27
|
Wang Z, Li X, Chen H, Han L, Ji X, Wang Q, Wei L, Miao Y, Wang J, Mao J, Zhang Z. Decreased HLF Expression Predicts Poor Survival in Lung Adenocarcinoma. Med Sci Monit 2021; 27:e929333. [PMID: 33979320 PMCID: PMC8127640 DOI: 10.12659/msm.929333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a type of non-small cell carcinoma. Its pathogenesis is being explored and there is no cure for the disease. Material/Methods The Gene Expression Omnibus (GEO) was searched to obtain data on expression of messenger RNA. GEO2R, an interactive web tool, was used to calculate the differentially expressed genes (DEGs) in LUAD. All the DEGs from different datasets were imported into VENNY 2.1 (https://bioinfogp.cnb.csic.es/tools/venny/index.html) to identify the intersection of the DEGs. An online analysis tool, the Database for Annotation, Visualization, and Integrated Discovery (DAVID), was used to help understand the biological meaning of DEG enrichment in LUAD. Cytoscape 3.7.2 was used to perform centrality analysis and visualize hub genes and related networks. Furthermore, the prognostic value of the hub genes was evaluated with the Kaplan-Meier plotter survival analysis tool. Results The GEO database was used to obtain RNA sequencing information for LUAD and normal tissue from the GSE118370, GSE136043, and GSE140797 datasets. A total of 376 DEGs were identified from GSE118370, 248 were identified from GSE136403, and 718 DEGs were identified from GSE140797. The 10 genes with the highest degrees of expression – the hub genes – were CAV1, TEK, SLIT2, RHOJ, DGSX, HLF, MEIS1, PTPRD, FOXF1, and ADRB2. In addition, Kaplan-Meier survival evaluation showed that CAV1, TEK, SLIT2, HLF, MEIS1, PTPRD, FOXF1, and ADRB2 were associated with favorable outcomes for LUAD. Conclusions CAV1, TEK, SLIT2, HLF, MEIS1, PTPRD, FOXF1, and ADRB2 are hub genes in the DEG interaction network for LUAD and are involved in the development of and prognosis for the disease. The mechanisms underlying these genes should be the subject of further studies.
Collapse
Affiliation(s)
- Zaiyan Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Xiaoning Li
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Hao Chen
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Li Han
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Xiaobin Ji
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Qiubo Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Li Wei
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Yafang Miao
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Jing Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Jianfeng Mao
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Zeming Zhang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| |
Collapse
|
28
|
Abstract
Prostate cancer is a global health problem, but incidence varies considerably across different continents. Asia is traditionally considered a low-incidence area, but the incidence and mortality of prostate cancer have rapidly increased across the continent. Substantial differences in epidemiological features have been observed among different Asian regions, and incidence, as well as mortality-to-incidence ratio, is associated with the human development index. Prostate cancer mortality decreased in Japan and Israel from 2007 to 2016, but mortality has increased in Thailand, Kyrgyzstan and Uzbekistan over the same period. Genomic analyses have shown a low prevalence of ERG oncoprotein in the East Asian population, alongside a low rate of PTEN loss, high CHD1 enrichments and high FOXA1 alterations. Contributions from single-nucleotide polymorphisms to prostate cancer risk vary with ethnicity, but germline mutation rates of DNA damage repair genes in metastatic prostate cancer are comparable in Chinese and white patients from the USA and UK. Pharmacogenomic features of testosterone metabolism might contribute to disparities seen in the response to androgen deprivation between East Asian men and white American and European men. Overall, considerable diversity in epidemiology and genomics of prostate cancer across Asia defines disease characteristics in these populations, but studies in this area are under-represented in the literature. Taking into account this intracontinental and intercontinental heterogeneity, translational studies are required in order to develop ethnicity-specific treatment strategies.
Collapse
|
29
|
Cheng S, Yang S, Shi Y, Shi R, Yeh Y, Yu X. Neuroendocrine prostate cancer has distinctive, non-prostatic HOX code that is represented by the loss of HOXB13 expression. Sci Rep 2021; 11:2778. [PMID: 33531604 PMCID: PMC7854582 DOI: 10.1038/s41598-021-82472-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/20/2021] [Indexed: 01/08/2023] Open
Abstract
HOX gene-encoded homeobox proteins control body patterning during embryonic development; the specific expression pattern of HOX genes may correspond to tissue identity. In this study, using RNAseq data of 1019 human cancer cell lines that originated from 24 different anatomic sites, we established HOX codes for various types of tissues. We applied these HOX codes to the transcriptomic profiles of prostate cancer (PCa) samples and found that the majority of prostate adenocarcinoma (AdPCa) samples sustained a prostate-specific HOX code whereas the majority of neuroendocrine prostate cancer (NEPCa) samples did not, which reflects the anaplastic nature of NEPCa. Also, our analysis showed that the NEPCa samples did not correlate well with the HOX codes of any other tissue types, indicating that NEPCa tumors lose their prostate identities but do not gain new tissue identities. Additionally, using immunohistochemical staining, we evaluated the prostatic expression of HOXB13, the most prominently changed HOX gene in NEPCa. We found that HOXB13 was expressed in both benign prostatic tissues and AdPCa but its expression was reduced or lost in NEPCa. Furthermore, we treated PCa cells with all trans retinoic acid (ATRA) and found that the reduced HOXB13 expression can be reverted. This suggests that ATRA is a potential therapeutic agent for the treatment of NEPCa tumors by reversing them to a more treatable AdPCa.
Collapse
Affiliation(s)
- Siyuan Cheng
- Department of Biochemistry and Molecular Biology, LSU Health-Shreveport, Shreveport, LA, USA
| | - Shu Yang
- Department of Biochemistry and Molecular Biology, LSU Health-Shreveport, Shreveport, LA, USA
| | - Yingli Shi
- Department of Biochemistry and Molecular Biology, LSU Health-Shreveport, Shreveport, LA, USA
| | - Runhua Shi
- Department of Medicine, LSU Health-Shreveport, Shreveport, LA, USA
| | - Yunshin Yeh
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical Center, Shreveport, LA, USA.,Department of Urology, LSU Health-Shreveport, Shreveport, LA, USA
| | - Xiuping Yu
- Department of Biochemistry and Molecular Biology, LSU Health-Shreveport, Shreveport, LA, USA. .,Department of Urology, LSU Health-Shreveport, Shreveport, LA, USA.
| |
Collapse
|
30
|
Jiang M, Xu S, Bai M, Zhang A. The emerging role of MEIS1 in cell proliferation and differentiation. Am J Physiol Cell Physiol 2020; 320:C264-C269. [PMID: 33296285 DOI: 10.1152/ajpcell.00422.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell proliferation and differentiation are the foundation of reproduction and growth. Mistakes in these processes may affect cell survival, or cause cell cycle dysregulation, such as tumorigenesis, birth defects and degenerative diseases, or cell death. Myeloid ecotropic viral integration site 1 (MEIS1) was initially discovered in leukemic mice. Recent research identified MEIS1 as an important transcription factor that regulates cell proliferation and differentiation during cell fate commitment. MEIS1 has a pro-proliferative effect in leukemia cells; however, its overexpression in cardiomyocytes restrains neonatal and adult cardiomyocyte proliferation. In addition, MEIS1 has carcinogenic or tumor suppressive effects in different neoplasms. Thus, this uncertainty suggests that MEIS1 has a unique function in cell proliferation and differentiation. In this review, we summarize the primary findings of MEIS1 in regulating cell proliferation and differentiation. Correlations between MEIS1 and cell fate specification might suggest MEIS1 as a therapeutic target for diseases.
Collapse
Affiliation(s)
- Mingzhu Jiang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, People's Republic of China
| | - Shuang Xu
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, People's Republic of China
| | - Mi Bai
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, People's Republic of China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Aihua Zhang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, People's Republic of China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
31
|
VanOpstall C, Perike S, Brechka H, Gillard M, Lamperis S, Zhu B, Brown R, Bhanvadia R, Vander Griend DJ. MEIS-mediated suppression of human prostate cancer growth and metastasis through HOXB13-dependent regulation of proteoglycans. eLife 2020; 9:e53600. [PMID: 32553107 PMCID: PMC7371429 DOI: 10.7554/elife.53600] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular roles of HOX transcriptional activity in human prostate epithelial cells remain unclear, impeding the implementation of new treatment strategies for cancer prevention and therapy. MEIS proteins are transcription factors that bind and direct HOX protein activity. MEIS proteins are putative tumor suppressors that are frequently silenced in aggressive forms of prostate cancer. Here we show that MEIS1 expression is sufficient to decrease proliferation and metastasis of prostate cancer cells in vitro and in vivo murine xenograft models. HOXB13 deletion demonstrates that the tumor-suppressive activity of MEIS1 is dependent on HOXB13. Integration of ChIP-seq and RNA-seq data revealed direct and HOXB13-dependent regulation of proteoglycans including decorin (DCN) as a mechanism of MEIS1-driven tumor suppression. These results define and underscore the importance of MEIS1-HOXB13 transcriptional regulation in suppressing prostate cancer progression and provide a mechanistic framework for the investigation of HOXB13 mutants and oncogenic cofactors when MEIS1/2 are silenced.
Collapse
Affiliation(s)
- Calvin VanOpstall
- The Committee on Cancer Biology, The University of ChicagoChicagoUnited States
| | - Srikanth Perike
- Department of Pathology, The University of Illinois at ChicagoChicagoUnited States
| | - Hannah Brechka
- The Committee on Cancer Biology, The University of ChicagoChicagoUnited States
| | - Marc Gillard
- Department of Surgery, Section of Urology, The University of ChicagoChicagoUnited States
| | - Sophia Lamperis
- Department of Pathology, The University of Illinois at ChicagoChicagoUnited States
| | - Baizhen Zhu
- Department of Surgery, Section of Urology, The University of ChicagoChicagoUnited States
| | - Ryan Brown
- Department of Pathology, The University of Illinois at ChicagoChicagoUnited States
| | - Raj Bhanvadia
- Department of Urology, UT SouthwesternDallasUnited States
| | | |
Collapse
|