1
|
Fang H, He J, Du D, Wang X, Xu X, Lu L, Zhou Y, Wen Y, He F, Li Y, Wen H, Zhou M. Deciphering the secret codes in N 7-methylguanosine modification: Context-dependent function of methyltransferase-like 1 in human diseases. Clin Transl Med 2025; 15:e70240. [PMID: 39979979 PMCID: PMC11842222 DOI: 10.1002/ctm2.70240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N7-methylguanosine (m7G) is one of the most prevalent post-transcriptional modifications of RNA and plays a critical role in RNA translation and stability. As a pivotal m7G regulator, methyltransferase-like 1 (METTL1) is responsible for methyl group transfer during the progression of m7G modification and contributes to the structure and functional regulation of RNA. Accumulating evidence in recent years has revealed that METTL1 plays key roles in various diseases depending on its m7G RNA methyltransferase activity. Elevated levels of METTL1 are typically associated with disease development and adverse consequences. In contrast, METTL1 may act as a disease suppressor in several disorders. While the roles of m7G modifications in disease have been extensively reviewed, the critical functions of METTL1 in various types of disease and the potential targeting of METTL1 for disease treatment have not yet been highlighted. This review describes the various biological functions of METTL1, summarises recent advances in understanding its pathogenic and disease-suppressive functions and discusses the underlying molecular mechanisms. Given that METTL1 can promote or inhibit disease processes, the possibility of applying METTL1 inhibitors and agonists is further discussed, with the goal of providing novel insights for future disease diagnosis and potential intervention targets. KEY POINTS: METTL1-mediated m7G modification is crucial for various biological processes, including RNA stability, maturation and translation. METTL1 has emerged as a critical epigenetic modulator in human illnesses, with its dysregulated expression correlating with multiple diseases progression and presenting opportunities for both diagnostic biomarker development and molecular-targeted therapy. Enormous knowledge gaps persist regarding context-dependent regulatory networks of METTL1 and dynamic m7G modification patterns, necessitating mechanistic interrogation to bridge basic research with clinical translation in precision medicine.
Collapse
Affiliation(s)
- Huan Fang
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jing He
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Dan Du
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xue Wang
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinyu Xu
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Linping Lu
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yefan Zhou
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yangyang Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Fucheng He
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yingxia Li
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongtao Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingxia Zhou
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
2
|
Samal S, Barik D, Shyamal S, Jena S, Panda AC, Dash M. Synergistic Interaction between Polysaccharide-Based Extracellular Matrix and Mineralized Osteoblast-Derived EVs Promotes Bone Regeneration via miRNA-mRNA Regulatory Axis. Biomacromolecules 2024; 25:4139-4155. [PMID: 38924768 DOI: 10.1021/acs.biomac.4c00269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Extracellular vesicles (EVs) derived from bone progenitor cells are advantageous as cell-free and non-immunogenic cargo delivery vehicles. In this study, EVs are isolated from MC3T3-E1 cells before (GM-EVs) and after mineralization for 7 and 14 days (DM-EVs). It was observed that DM-EVs accelerate the process of differentiation in recipient cells more prominently. The small RNA sequencing of EVs revealed that miR-204-5p, miR-221-3p, and miR-148a-3p are among the highly upregulated miRNAs that have an inhibitory effect on the function of mRNAs, Sox11, Timp3, and Ccna2 in host cells, which is probably responsible for enhancing the activity of osteoblastic genes. To enhance the bioavailability of EVs, they are encapsulated in a chitosan-collagen composite hydrogel that serves as a bioresorbable extracellular matrix (ECM). The EVs-integrated scaffold (DM-EVs + Scaffold) enhances bone regeneration in critical-sized calvarial bone defects in rats within 8 weeks of implantation by providing the ECM cues. The shelf life of DM-EVs + Scaffold indicates that the bioactivity of EVs and their cargo in the polymer matrix remains intact for up to 30 days. Integrating mineralized cell-derived EVs into an ECM represents a bioresorbable matrix with a cell-free method for promoting new bone formation through the miRNA-mRNA regulatory axis.
Collapse
Affiliation(s)
- Sasmita Samal
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024 Odisha, India
| | - Debyashreeta Barik
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024 Odisha, India
| | - Sharmishtha Shyamal
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- ICMR-National Institute for Reproduction Biology and Child Health, Mumbai 400012, India
| | - Sarita Jena
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| | - Amaresh C Panda
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| | - Mamoni Dash
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| |
Collapse
|
3
|
Russo S, Kwiatkowski M, Wolters JC, Gerding A, Hermans J, Govorukhina N, Bischoff R, Melgert BN. Effects of lysine deacetylase inhibitor treatment on LPS responses of alveolar-like macrophages. J Leukoc Biol 2024; 115:435-449. [PMID: 37811856 DOI: 10.1093/jleuko/qiad121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages are key immune cells that can adapt their metabolic phenotype in response to different stimuli. Lysine deacetylases are important enzymes regulating inflammatory gene expression and lysine deacetylase inhibitors have been shown to exert anti-inflammatory effects in models of chronic obstructive pulmonary disease. We hypothesized that these anti-inflammatory effects may be associated with metabolic changes in macrophages. To validate this hypothesis, we used an unbiased and a targeted proteomic approach to investigate metabolic enzymes, as well as liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry, to quantify metabolites in combination with the measurement of functional parameters in primary murine alveolar-like macrophages after lipopolysaccharide-induced activation in the presence or absence of lysine deacetylase inhibition. We found that lysine deacetylase inhibition resulted in reduced production of inflammatory mediators such as tumor necrosis factor α and interleukin 1β. However, only minor changes in macrophage metabolism were observed, as only one of the lysine deacetylase inhibitors slightly increased mitochondrial respiration while no changes in metabolite levels were seen. However, lysine deacetylase inhibition specifically enhanced expression of proteins involved in ubiquitination, which may be a driver of the anti-inflammatory effects of lysine deacetylase inhibitors. Our data illustrate that a multiomics approach provides novel insights into how macrophages interact with cues from their environment. More detailed studies investigating ubiquitination as a potential driver of lysine deacetylase inhibition will help developing novel anti-inflammatory drugs for difficult-to-treat diseases such as chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Sara Russo
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Marcel Kwiatkowski
- Functional Proteo-Metabolomics, Department of Biochemistry, University of Innsbruck, Innrain 80-82, Innsbruck 6020, Austria
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Albert Gerding
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Jos Hermans
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Natalia Govorukhina
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Barbro N Melgert
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| |
Collapse
|
4
|
Chen XZ, Li XM, Xu SJ, Hu S, Wang T, Li RF, Liu CY, Xue JQ, Zhou LY, Wang YH, Li PF, Wang K. TMEM11 regulates cardiomyocyte proliferation and cardiac repair via METTL1-mediated m 7G methylation of ATF5 mRNA. Cell Death Differ 2023:10.1038/s41418-023-01179-0. [PMID: 37286744 DOI: 10.1038/s41418-023-01179-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/19/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
The mitochondrial transmembrane (TMEM) protein family has several essential physiological functions. However, its roles in cardiomyocyte proliferation and cardiac regeneration remain unclear. Here, we detected that TMEM11 inhibits cardiomyocyte proliferation and cardiac regeneration in vitro. TMEM11 deletion enhanced cardiomyocyte proliferation and restored heart function after myocardial injury. In contrast, TMEM11-overexpression inhibited neonatal cardiomyocyte proliferation and regeneration in mouse hearts. TMEM11 directly interacted with METTL1 and enhanced m7G methylation of Atf5 mRNA, thereby increasing ATF5 expression. A TMEM11-dependent increase in ATF5 promoted the transcription of Inca1, an inhibitor of cyclin-dependent kinase interacting with cyclin A1, which suppressed cardiomyocyte proliferation. Hence, our findings revealed that TMEM11-mediated m7G methylation is involved in the regulation of cardiomyocyte proliferation, and targeting the TMEM11-METTL1-ATF5-INCA1 axis may serve as a novel therapeutic strategy for promoting cardiac repair and regeneration.
Collapse
Affiliation(s)
- Xin-Zhe Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xin-Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Shi-Jun Xu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029, Beijing, China
| | - Shen Hu
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology, 100730, Beijing, China
| | - Tao Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Rui-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cui-Yun Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Jun-Qiang Xue
- Department of Rehabilitation Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lu-Yu Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yun-Hong Wang
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 100037, Beijing, China.
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
5
|
Zhang H, Qian J, Jin M, Fan L, Fan S, Pan H, Li Y, Wang N, Jian B. Jolkinolide B induces cell cycle arrest and apoptosis in MKN45 gastric cancer cells and inhibits xenograft tumor growth in vivo. Biosci Rep 2022; 42:BSR20220341. [PMID: 35674158 PMCID: PMC9245080 DOI: 10.1042/bsr20220341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Gastric cancer is one of the most common digestive carcinomas throughout the world and represents high mortality. There is an urgent quest for seeking a novel and efficient antigastric cancer drug. Euphorbia fischeriana Steud had long been used as a traditional Chinese medicine for the treatment of cancer. According to the basic theory of traditional Chinese medicine, its antitumor mechanism is 'to combat poison with poison'. However, its effective material foundation of it is still ambiguous. In our previous work, we studied the chemical constituents of E. fischeriana Steud. Jolkinolide B (JB) is an ent-abietane-type diterpenoid we isolated from it. The purpose of the present study was to investigate the antigastric effect and mechanism of JB. Results revealed that JB could suppress the proliferation of MKN45 cells in vitro and inhibit MKN45 xenograft tumor growth in nude mice in vivo. We further investigated its anticancer mechanism. On the one hand, JB caused DNA damage in gastric cancer MKN45 cells and induced the S cycle arrest by activating the ATR-CHK1-CDC25A-Cdk2 signaling pathway, On the other hand, JB induced MKN45 cells apoptosis through the mitochondrial pathway, and ultimately effectively inhibited the growth of gastric cancer cells. These results suggest that JB appears to be a promising candidate drug with antigastric cancer activity and warrants further research.
Collapse
Affiliation(s)
- Hao Zhang
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Jiayi Qian
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Ming Jin
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Li Fan
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - SongJie Fan
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Hong Pan
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Yang Li
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Ningning Wang
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| | - Baiyu Jian
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161000, P. R. China
| |
Collapse
|
6
|
ElGendy K, Malcomson FC, Bradburn DM, Mathers JC. Effects of bariatric surgery on DNA methylation in adults: a systematic review and meta-analysis. Surg Obes Relat Dis 2019; 16:128-136. [PMID: 31708383 DOI: 10.1016/j.soard.2019.09.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND DNA methylation is an epigenetic mechanism through which environmental factors, including obesity, influence health. Obesity is a major modifiable risk factor for many common diseases, including cardiovascular diseases and cancer. Obesity-induced metabolic stress and inflammation are key mechanisms that affect disease risk and that may result from changes in methylation of metabolic and inflammatory genes. OBJECTIVES This review aims to report the effects of weight loss induced by bariatric surgery (BS) on DNA methylation in adults with obesity focusing on changes in metabolic and inflammatory genes. METHODS A systematic review was performed using MEDLINE, EMBASE, and Scopus, to identify studies in adult humans that reported DNA methylation after BS. RESULTS Of 15,996 screened titles, 15 intervention studies were identified, all of which reported significantly lower body mass index postsurgery. DNA methylation was assessed in 5 different tissues (blood = 7 studies, adipose tissues = 4, skeletal muscle = 2, liver, and spermatozoa). Twelve studies reported significant changes in DNA methylation after BS. Meta-analysis showed that BS increased methylation of PDK4 loci in skeletal muscle and blood in 2 studies, while the effects of BS on IL6 methylation levels in blood were inconsistent. BS had no overall effect on LINE1 or PPARGC1 methylation. CONCLUSION The current evidence supports the reversibility of DNA methylation at specific loci in response to BS-induced weight loss. These changes are consistent with improved metabolic and inflammatory profiles of patients after BS. However, the evidence regarding the effects of BS on DNA methylation in humans is limited and inconsistent, which makes it difficult to combine and compare data across studies.
Collapse
Affiliation(s)
- Khalil ElGendy
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Surgery Department, Northumbria NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| | - Fiona C Malcomson
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - D Michael Bradburn
- Surgery Department, Northumbria NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
7
|
Ma HT, Niu CM, Xia J, Shen XY, Xia MM, Hu YQ, Zheng Y. Stimulated by retinoic acid gene 8 (Stra8) plays important roles in many stages of spermatogenesis. Asian J Androl 2019; 20:479-487. [PMID: 29848833 PMCID: PMC6116687 DOI: 10.4103/aja.aja_26_18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
To clarify the functions and mechanism of stimulated by retinoic acid gene 8 (Stra8) in spermatogenesis, we analyzed the testes from Stra8 knockout and wild-type mice during the first wave of spermatogenesis. Comparisons showed no significant differences in morphology and number of germ cells at 11 days postpartum, while 21 differentially expressed genes (DEGs) associated with spermatogenesis were identified. We speculate that Stra8 performs many functions in different phases of spermatogenesis, such as establishment of spermatogonial stem cells, spermatogonial proliferation and self-renewal, spermatogonial differentiation and meiosis, through direct or indirect regulation of these DEGs. We therefore established a preliminary regulatory network of Stra8 during spermatogenesis. These results will provide a theoretical basis for further research on the mechanism underlying the role of Stra8 in spermatogenesis.
Collapse
Affiliation(s)
- Hai-Tao Ma
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| | - Chang-Min Niu
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| | - Jing Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| | - Xue-Yi Shen
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| | - Meng-Meng Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| | - Yan-Qiu Hu
- Clinicial Medical College, Yangzhou University, Yangzhou 225001, China
| | - Ying Zheng
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou 225001, China.,Jiangsu Key Laboratory of Experimental and Translational Noncoding RNA Research, Yangzhou 225001, China
| |
Collapse
|
8
|
Zhang L, Cao H, He T, Yang J, Tao H, Wang Y, Hu Q. Overexpression of PRDM13 inhibits glioma cells via Rho and GTP enzyme activation protein. Int J Mol Med 2018; 42:966-974. [PMID: 29767251 PMCID: PMC6034930 DOI: 10.3892/ijmm.2018.3679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/10/2018] [Indexed: 11/17/2022] Open
Abstract
PR (PRDI-BFI and RIZ) domain containing (PRDM) proteins have been shown to be important in several types of human cancer. PRDM13, a member of the PRDM family, contains transcriptional regulators involved in modulating several cellular processes. However, the function of PRDM13 in glioma remains to be elucidated. The purpose of the present study was to evaluate the expression and effect of PRDM13 on glioma cells. It was found that the expression of PRDM13 was reduced in glioma cells, and the overexpression of PRDM13 significantly decreased the proliferation, migration and invasion of U87 glioma cells. Through validation of RNA-sequencing analysis, genes regulating cell proliferation and migration were classified from Gene Ontology sources. In addition, PRDM13 was shown to be associated with Rho protein and GTP enzyme activation protein. The over expression of PRDM13 upregulated deleted in liver cancer 1 (DLC1) to inhibit the proliferation and invasion of U87 cells. In conclusion, PRDM13 decreased the proliferation and invasion of U87 cells, and may be of potential value for glioma therapy.
Collapse
Affiliation(s)
- Linna Zhang
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Huimei Cao
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao He
- Department of Spinal Surgery, Dongfeng General Hospital of Chinese Medicine Affiliated to Hubei Medical University, Shiyan, Hubei 442000, P.R. China
| | - Jijuan Yang
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hong Tao
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yin Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Basic Medical School of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qikuan Hu
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
9
|
Coppin E, Florentin J, Vasamsetti SB, Arunkumar A, Sembrat J, Rojas M, Dutta P. Splenic hematopoietic stem cells display a pre-activated phenotype. Immunol Cell Biol 2018; 96:10.1111/imcb.12035. [PMID: 29526053 PMCID: PMC6379147 DOI: 10.1111/imcb.12035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/16/2018] [Accepted: 03/05/2018] [Indexed: 12/12/2022]
Abstract
Splenic hematopoiesis is crucial to the pathogenesis of diseases including myocardial infarction and atherosclerosis. The spleen acts as a reservoir of myeloid cells, which are quickly expelled out in response to acute inflammation. In contrast to the well-defined bone marrow hematopoiesis, the cellular and molecular components sustaining splenic hematopoiesis are poorly understood. Surprisingly, we found that, unlike quiescent bone marrow hematopoietic stem cells (HSC), most of splenic HSC are in the G1 phase in C57BL/6 mice. Moreover, splenic HSC were enriched for genes involved in G0-G1 transition and expressed lower levels of genes responsible for G1-S transition. These data indicate that, at steady state, splenic HSC are pre-activated, which may expedite their cell cycle entry in emergency conditions. Consistently, in the acute phase of septic shock induced by LPS injection, splenic HSC entered the S-G2-M phase, whereas bone marrow HSC did not. Mobilization and transplantation experiments displayed that bone marrow HSC, once in the spleen, acquired cell cycle status similar to splenic HSC, strongly suggesting that the splenic microenvironment plays an important role in HSC pre-activation. In addition, we found that myeloid translocation gene 16 (Mtg16) deficiency in C57BL/6 mice resulted in significantly increased S-G2-M entry of splenic but not bone marrow HSC, suggesting that Mtg16 is an intrinsic negative regulator of G1-S transition in splenic HSC. Altogether, this study demonstrates that compared to bone marrow, splenic HSC are in a pre-activated state, which is driven by extracellular signals provided by splenic microenvironment and HSC intrinsic factor Mtg16.
Collapse
Affiliation(s)
- Emilie Coppin
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Jonathan Florentin
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sathish Babu Vasamsetti
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Anagha Arunkumar
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - John Sembrat
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, PA 15261, USA
| | - Mauricio Rojas
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, PA 15261, USA
| | - Partha Dutta
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
10
|
Gallitz I, Lofruthe N, Traeger L, Bäumer N, Hoerr V, Faber C, Kuhlmann T, Müller-Tidow C, Steinbicker AU. Deficiency of the BMP Type I receptor ALK3 partly protects mice from anemia of inflammation. BMC PHYSIOLOGY 2018; 18:3. [PMID: 29482530 PMCID: PMC6389079 DOI: 10.1186/s12899-018-0037-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
Abstract
Background Inflammatory stimuli induce the hepatic iron regulatory hormone hepcidin, which contributes to anaemia of inflammation (AI). Hepcidin expression is regulated by the bone morphogenetic protein (BMP) and the interleukin-6 (IL-6) signalling pathways. Prior results indicate that the BMP type I receptor ALK3 is mainly involved in the acute inflammatory hepcidin induction four and 72 h after IL-6 administration. In this study, the role of ALK3 in a chronic model of inflammation was investigated. The intact, heat-killed bacterium Brucella abortus (BA) was used to analyse its effect on the development of inflammation and hypoferremia in mice with hepatocyte-specific Alk3-deficiency (Alk3fl/fl; Alb-Cre) compared to control (Alk3fl/fl) mice. Results An iron restricted diet prevented development of the iron overload phenotype in mice with hepatocyte-specific Alk3 deficiency. Regular diet leads to iron overload and increased haemoglobin levels in these mice, which protects from the development of AI per se. Fourteen days after BA injection Alk3fl/fl; Alb-Cre mice presented milder anaemia (Hb 16.7 g/dl to 11.6 g/dl) compared to Alk3fl/fl control mice (Hb 14.9 g/dl to 8.6 g/dl). BA injection led to an intact inflammatory response in all groups of mice. In Alk3fl/fl; Alb-Cre mice, SMAD1/5/8 phosphorylation was reduced after BA as well as after infection with Staphylococcus aureus. The reduction of the SMAD1/5/8 signalling pathway due to hepatocyte-specific Alk3 deficiency partly suppressed the induction of STAT3 signalling. Conclusion The results reveal in vivo, that 1) hepatocyte-specific Alk3 deficiency partly protects from AI, 2) the development of hypoferremia is partly dependent on ALK3, and 3) the ALK3/BMP/hepcidin axis may serve as a possible therapeutic target to attenuate AI. Electronic supplementary material The online version of this article (10.1186/s12899-018-0037-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Inka Gallitz
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Niklas Lofruthe
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Lisa Traeger
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Nicole Bäumer
- Department of Medicine A, Molecular Haematology and Oncology, University Hospital Muenster, 48149, Muenster, Germany
| | - Verena Hoerr
- Institute of Medical Microbiology, Jena University Hospital, 07747, Jena, Germany.,Department of Clinical Radiology, University Hospital Muenster, 48149, Muenster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Muenster, 48149, Muenster, Germany
| | - Tanja Kuhlmann
- Institute for Neuropathology, University Hospital Muenster, 48149, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine A, Molecular Haematology and Oncology, University Hospital Muenster, 48149, Muenster, Germany.,Present Address: Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Andrea U Steinbicker
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany.
| |
Collapse
|
11
|
Kim TH, Park JM, Kim MY, Ahn YH. The role of CREB3L4 in the proliferation of prostate cancer cells. Sci Rep 2017; 7:45300. [PMID: 28338058 PMCID: PMC5364418 DOI: 10.1038/srep45300] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/23/2017] [Indexed: 12/19/2022] Open
Abstract
The incidence of prostate cancer (PC) is growing rapidly throughout the world, in probable association with the adoption of western style diets. Thus, understanding the molecular pathways triggering the development of PC is crucial for both its prevention and treatment. Here, we investigated the role of the metabolism-associated protein, CREB3L4, in the proliferation of PC cells. CREB3L4 was upregulated by the synthetic androgen, R1881, in LNCaP PC cells (an androgen-dependent cell line). Knockdown of CREB3L4 resulted in decreased androgen-dependent PC cell growth. LNCaP cells transfected with siCREB3L4 underwent G2/M arrest, with upregulation of the proteins cyclin B1, phospho-CDK1, p21Waf1/Cip1, and INCA1, and downregulation of cyclin D1. Moreover, depletion of CREB3L4 resulted in significantly decreased expression of a subset of androgen-receptor (AR) target genes, including PSA, FKBP5, HPGD, KLK2, and KLK4. We also demonstrated that CREB3L4 directly interacts with the AR, and increases the binding of AR to androgen response elements (AREs). We also identified a role for the unfolded protein response (and its surrogate, IRE1α), in activating CREB3L4. Cumulatively, we postulate that CREB3L4 expression is mediated by an AR-IRE1α axis, but is also directly regulated by AR-to-ARE binding. Thus, our study demonstrates that CREB3L4 plays a key role in PC cell proliferation, which is promoted by both AR and IRE1α.
Collapse
Affiliation(s)
- Tae-Hyun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Joo-Man Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Mi-Young Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Yong-Ho Ahn
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| |
Collapse
|
12
|
Lofruthe N, Gallitz I, Traeger L, Bäumer N, Schulze I, Kuhlmann T, Müller-Tidow C, Steinbicker AU. Intravenous Iron Carboxymaltose as a Potential Therapeutic in Anemia of Inflammation. PLoS One 2016; 11:e0158599. [PMID: 27404499 PMCID: PMC4942094 DOI: 10.1371/journal.pone.0158599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/18/2016] [Indexed: 01/28/2023] Open
Abstract
Intravenous iron supplementation is an effective therapy in iron deficiency anemia (IDA), but controversial in anemia of inflammation (AI). Unbound iron can be used by bacteria and viruses for their replication and enhance the inflammatory response. Nowadays available high molecular weight iron complexes for intravenous iron substitution, such as ferric carboxymaltose, might be useful in AI, as these pharmaceuticals deliver low doses of free iron over a prolonged period of time. We tested the effects of intravenous iron carboxymaltose in murine AI: Wild-type mice were exposed to the heat-killed Brucella abortus (BA) model and treated with or without high molecular weight intravenous iron. 4h after BA injection followed by 2h after intravenous iron treatment, inflammatory cytokines were upregulated by BA, but not enhanced by iron treatment. In long term experiments, mice were fed a regular or an iron deficient diet and then treated with intravenous iron or saline 14 days after BA injection. Iron treatment in mice with BA-induced AI was effective 24h after iron administration. In contrast, mice with IDA (on iron deficiency diet) prior to BA-IA required 7d to recover from AI. In these experiments, inflammatory markers were not further induced in iron-treated compared to vehicle-treated BA-injected mice. These results demonstrate that intravenous iron supplementation effectively treated the murine BA-induced AI without further enhancement of the inflammatory response. Studies in humans have to reveal treatment options for AI in patients.
Collapse
Affiliation(s)
- Niklas Lofruthe
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Inka Gallitz
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Lisa Traeger
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Nicole Bäumer
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Isabell Schulze
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Tanja Kuhlmann
- Institute for Neuropathology, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Andrea U. Steinbicker
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
13
|
Bäumer N, Bäumer S, Haak M, Koschmieder S, Schönig K, Berdel WE, Müller-Tidow C. A Limited Role for the Cell Cycle Regulator Cyclin A1 in Murine Leukemogenesis. PLoS One 2015; 10:e0129147. [PMID: 26080083 PMCID: PMC4469679 DOI: 10.1371/journal.pone.0129147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/05/2015] [Indexed: 12/04/2022] Open
Abstract
The quest for novel therapeutic targets in acute myeloid leukemia (AML) is still ongoing. One of such targets, cyclin A1, was shown to be overexpressed in AML including AML stem cells. However, the function of cyclin A1 in AML is largely unknown, and the data on its impact on patients´ survival remain controversial. Therefore, we developed a transgenic mouse model of stem cell-directed inducible cyclin A1 overexpression and crossed these mice with PML-RARα-knockin mice, which develop an AML M3-like phenotype. To observe the effects of cyclin A1 loss-of-function, we also crossed PML-RARα-knockin mice to cyclin A1-knockout mice. Neither overexpression nor loss of cyclin A1 significantly altered leukemogenesis in PML-RARα-knockin mice. These findings imply that upregulation of cyclin A1 is not essential for leukemogenesis. Our data suggest that cyclin A1 does not represent a suitable target for AML therapy.
Collapse
Affiliation(s)
- Nicole Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- * E-mail: (NB); (CMT)
| | - Sebastian Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Miriam Haak
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Steffen Koschmieder
- Department of Medicine (Hematology, Oncology, Hemostaseology, and SCT), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Kai Schönig
- Central Institute of Mental Health, Department of Molecular Biology, Heidelberg University, Mannheim, Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Dept. of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
- * E-mail: (NB); (CMT)
| |
Collapse
|
14
|
Nilsson EK, Ernst B, Voisin S, Almén MS, Benedict C, Mwinyi J, Fredriksson R, Schultes B, Schiöth HB. Roux-en Y gastric bypass surgery induces genome-wide promoter-specific changes in DNA methylation in whole blood of obese patients. PLoS One 2015; 10:e0115186. [PMID: 25710379 PMCID: PMC4340013 DOI: 10.1371/journal.pone.0115186] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
CONTEXT DNA methylation has been proposed to play a critical role in many cellular and biological processes. OBJECTIVE To examine the influence of Roux-en-Y gastric bypass (RYGB) surgery on genome-wide promoter-specific DNA methylation in obese patients. Promoters are involved in the initiation and regulation of gene transcription. METHODS Promoter-specific DNA methylation in whole blood was measured in 11 obese patients (presurgery BMI >35 kg/m(2), 4 females), both before and 6 months after RYGB surgery, as well as once only in a control group of 16 normal-weight men. In addition, body weight and fasting plasma glucose were measured after an overnight fast. RESULTS The mean genome-wide distance between promoter-specific DNA methylation of obese patients at six months after RYGB surgery and controls was shorter, as compared to that at baseline (p<0.001). Moreover, postsurgically, the DNA methylation of 51 promoters was significantly different from corresponding values that had been measured at baseline (28 upregulated and 23 downregulated, P<0.05 for all promoters, Bonferroni corrected). Among these promoters, an enrichment for genes involved in metabolic processes was found (n = 36, P<0.05). In addition, the mean DNA methylation of these 51 promoters was more similar after surgery to that of controls, than it had been at baseline (P<0.0001). When controlling for the RYGB surgery-induced drop in weight (-24% of respective baseline value) and fasting plasma glucose concentration (-16% of respective baseline value), the DNA methylation of only one out of 51 promoters (~2%) remained significantly different between the pre-and postsurgery time points. CONCLUSIONS Epigenetic modifications are proposed to play an important role in the development of and predisposition to metabolic diseases, including type II diabetes and obesity. Thus, our findings may form the basis for further investigations to unravel the molecular effects of gastric bypass surgery. CLINICAL TRIAL ClinicalTrials.gov NCT01730742.
Collapse
Affiliation(s)
- Emil K. Nilsson
- Department of Neuroscience, BMC, box 593, 75124 Uppsala, Sweden
- * E-mail:
| | - Barbara Ernst
- Interdisciplinary Obesity Center, eSwiss Medical & Surgical Center, St. Gallen, Switzerland
| | - Sarah Voisin
- Department of Neuroscience, BMC, box 593, 75124 Uppsala, Sweden
| | | | | | - Jessica Mwinyi
- Department of Neuroscience, BMC, box 593, 75124 Uppsala, Sweden
| | - Robert Fredriksson
- Interdisciplinary Obesity Center, eSwiss Medical & Surgical Center, St. Gallen, Switzerland
| | - Bernd Schultes
- Interdisciplinary Obesity Center, eSwiss Medical & Surgical Center, St. Gallen, Switzerland
| | | |
Collapse
|
15
|
Maintenance of leukemia-initiating cells is regulated by the CDK inhibitor Inca1. PLoS One 2014; 9:e115578. [PMID: 25525809 PMCID: PMC4272264 DOI: 10.1371/journal.pone.0115578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/01/2014] [Indexed: 11/19/2022] Open
Abstract
Functional differences between healthy progenitor and cancer initiating cells may provide unique opportunities for targeted therapy approaches. Hematopoietic stem cells are tightly controlled by a network of CDK inhibitors that govern proliferation and prevent stem cell exhaustion. Loss of Inca1 led to an increased number of short-term hematopoietic stem cells in older mice, but Inca1 seems largely dispensable for normal hematopoiesis. On the other hand, Inca1-deficiency enhanced cell cycling upon cytotoxic stress and accelerated bone marrow exhaustion. Moreover, AML1-ETO9a-induced proliferation was not sustained in Inca1-deficient cells in vivo. As a consequence, leukemia induction and leukemia maintenance were severely impaired in Inca1−/− bone marrow cells. The re-initiation of leukemia was also significantly inhibited in absence of Inca1−/− in MLL—AF9- and c-myc/BCL2-positive leukemia mouse models. These findings indicate distinct functional properties of Inca1 in normal hematopoietic cells compared to leukemia initiating cells. Such functional differences might be used to design specific therapy approaches in leukemia.
Collapse
|
16
|
Bäumer N, Krause A, Köhler G, Lettermann S, Evers G, Hascher A, Bäumer S, Berdel WE, Müller-Tidow C, Tickenbrock L. Proteinase-Activated Receptor 1 (PAR1) regulates leukemic stem cell functions. PLoS One 2014; 9:e94993. [PMID: 24740120 PMCID: PMC3989293 DOI: 10.1371/journal.pone.0094993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/21/2014] [Indexed: 12/30/2022] Open
Abstract
External signals that are mediated by specific receptors determine stem cell fate. The thrombin receptor PAR1 plays an important role in haemostasis, thrombosis and vascular biology, but also in tumor biology and angiogenesis. Its expression and function in hematopoietic stem cells is largely unknown. Here, we analyzed expression and function of PAR1 in primary hematopoietic cells and their leukemic counterparts. AML patients' blast cells expressed much lower levels of PAR1 mRNA and protein than CD34+ progenitor cells. Constitutive Par1-deficiency in adult mice did not affect engraftment or stem cell potential of hematopoietic cells. To model an AML with Par1-deficiency, we retrovirally introduced the oncogene MLL-AF9 in wild type and Par1−/− hematopoietic progenitor cells. Par1-deficiency did not alter initial leukemia development. However, the loss of Par1 enhanced leukemic stem cell function in vitro and in vivo. Re-expression of PAR1 in Par1−/− leukemic stem cells delayed leukemogenesis in vivo. These data indicate that Par1 contributes to leukemic stem cell maintenance.
Collapse
Affiliation(s)
- Nicole Bäumer
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Annika Krause
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Gabriele Köhler
- Gerhard Domagk Institute for Pathology, University of Muenster, Muenster, Germany
| | - Stephanie Lettermann
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Georg Evers
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Antje Hascher
- Hochschule Hamm-Lippstadt, University of Applied Science, Hamm, Germany
| | - Sebastian Bäumer
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Wolfgang E. Berdel
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
- Interdisciplinary Center for Clinical Research IZKF, University of Muenster, Muenster, Germany
- Dept. of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
- * E-mail: (CMT); (LT)
| | - Lara Tickenbrock
- Department of Medicine, Hematology/Oncology, University of Muenster, Muenster, Germany
- Hochschule Hamm-Lippstadt, University of Applied Science, Hamm, Germany
- * E-mail: (CMT); (LT)
| |
Collapse
|
17
|
Freitas MJ, Korrodi-Gregório L, Morais-Santos F, Cruz e Silva ED, Fardilha M. TCTEX1D4 interactome in human testis: unraveling the function of dynein light chain in spermatozoa. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:242-53. [PMID: 24606217 DOI: 10.1089/omi.2013.0133] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Studies were designed to identify the TCTEX1D4 interactome in human testis, with the purpose of unraveling putative protein complexes essential to male reproduction and thus novel TCTEX1D4 functions. TCTEX1D4 is a dynein light chain that belongs to the DYNT1/TCTEX1 family. In spermatozoa, it appears to be important to sperm motility, intraflagellar transport, and acrosome reaction. To contribute to the knowledge on TCTEX1D4 function in testis and spermatozoa, a yeast two-hybrid assay was performed in testis, which allowed the identification of 40 novel TCTEX1D4 interactors. Curiously, another dynein light chain, TCTEX1D2, was identified and its existence demonstrated for the first time in human spermatozoa. Immunofluorescence studies proved that TCTEX1D2 is an intra-acrosomal protein also present in the midpiece, suggesting a role in cargo movement in human spermatozoa. Further, an in silico profile of TCTEX1D4 revealed that most TCTEX1D4 interacting proteins were not previously characterized and the ones described present a very broad nature. This reinforces TCTEX1D4 as a dynein light chain that is capable of interacting with a variety of functionally different proteins. These observations collectively contribute to a deeper molecular understanding of the human spermatozoa function.
Collapse
Affiliation(s)
- Maria João Freitas
- Signal Transduction Laboratory, Centre for Cell Biology, Biology Department, Health Sciences Department, University of Aveiro , Aveiro, Portugal
| | | | | | | | | |
Collapse
|
18
|
Zhang F, Bäumer N, Rode M, Ji P, Zhang T, Berdel WE, Müller-Tidow C. The inhibitor of growth protein 5 (ING5) depends on INCA1 as a co-factor for its antiproliferative effects. PLoS One 2011; 6:e21505. [PMID: 21750715 PMCID: PMC3130024 DOI: 10.1371/journal.pone.0021505] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 05/30/2011] [Indexed: 12/25/2022] Open
Abstract
The proteins of the Inhibitor of Growth (ING) family are involved in multiple cellular functions such as cell cycle regulation, apoptosis, and chromatin remodeling. For ING5, its actual role in growth suppression and the necessary partners are not known. In a yeast-two-hybrid approach with human bone marrow derived cDNA, we identified ING5 as well as several other proteins as interaction partners of Inhibitor of cyclin A1 (INCA1) that we previously characterized as a novel interaction partner of cyclin A1/CDK2. ING5 expression in leukemic AML blasts was severely reduced compared to normal bone marrow. In line, ING5 inhibited bone marrow colony formation upon retroviral transduction. However, Inca1(-/-) bone marrow colony formation was not suppressed by ING5. In murine embryonic fibroblast (MEF) cells from Inca1(+/+) and Inca1(-/-) mice, overexpression of ING5 suppressed cell proliferation only in the presence of INCA1, while ING5 had no effect in Inca1(-/-) MEFs. ING5 overexpression induced a delay in S-phase progression, which required INCA1. Finally, ING5 overexpression enhanced Fas-induced apoptosis in Inca1(+/+) MEFs, while Inca1(-/-) MEFs were protected from Fas antibody-induced apoptosis. Taken together, these results indicate that ING5 is a growth suppressor with suppressed expression in AML whose functions depend on its interaction with INCA1.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi'an, China
| | - Nicole Bäumer
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
| | - Miriam Rode
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
| | - Ping Ji
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
- Department of Pathology, MD Anderson Cancer Center, Texas University, Houston, Texas, United States of America
| | - Tao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Wolfgang E. Berdel
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology/Oncology, University of Münster, Münster, Germany
- * E-mail:
| |
Collapse
|