1
|
Song X, Xu Y, Li M, Guan X, Liu H, Zhang J, Sun H, Ma C, Zhang L, Zhao X, Zheng X, Zhu D. SRSF4-Associated ca-circFOXP1 Regulates Hypoxia-Induced PASMC Proliferation by the Formation of R Loop With Host Gene. Arterioscler Thromb Vasc Biol 2025; 45:e118-e135. [PMID: 39973750 DOI: 10.1161/atvbaha.124.322026] [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: 11/14/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a rare and fatal disease, the pathological changes of which include pulmonary arterial smooth muscle cell (PASMC) proliferation, which is the pathological basis of pulmonary vascular remodeling. Studies have demonstrated that chromatin-associated circRNA can regulate a variety of biological processes. However, the role of chromatin-associated circRNA in the proliferation of PH remains largely unexplored. In this study, we aimed to identify the function and mechanism of chromatin-associated circRNA in PASMC proliferation in PH. METHODS The role of chromatin-associated circFOXP1 (ca-circFOXP1) was investigated in hypoxic mouse PASMCs and SuHX (Sugen5416+hypoxia) model mice through the use of antisense oligonucleotide knockdown and adeno-associated virus-mediated knockdown. Through bioinformatic sequence alignment, chromatin isolation by RNA purification, Cell Counting Kit 8, 5-ethynyl-2-deoxyuridine, Western blot, and other experiments, the function and mechanism of ca-circFOXP1 were verified. RESULTS The expression of ca-circFOXP1 was found to be significantly increased in SuHX model mice and hypoxic mouse PASMCs. Moreover, ca-circFOXP1 was found to regulate the level of the host protein FOXP1 (forkhead box protein 1) through the R loop, thereby influencing the phosphorylation activity of SMAD2 (SMAD family member 2) and, consequently, the proliferation of mouse PASMCs. It is noteworthy that the m6A modification was found to promote the formation of the R loop between ca-circFOXP1 and the host gene FOXP1, thereby regulating the expression of the host protein. Furthermore, we have identified that the splicing factor SRSF4 (serine/arginine rich splicing factor 4) can upregulate the expression of ca-circFOXP1 by splicing exons 6 and 9 of FOXP1 pre-mRNA. CONCLUSIONS The results demonstrated that the splicing factor SRSF4 upregulated the expression of ca-circFOXP1, and m6A methylation promoted R-loop formation between ca-circFOXP1 and host genes, regulated the level of host protein FOXP1, and then affected the phosphorylation activity of SMAD2, mediating PASMC proliferation, leading to pulmonary vascular remodeling. These results provide a theoretical basis for further study of the pathological mechanisms of hypoxic PH and may provide certain insights.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Mice
- Serine-Arginine Splicing Factors/metabolism
- Serine-Arginine Splicing Factors/genetics
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Male
- Smad2 Protein/metabolism
- Cell Hypoxia
- Mice, Inbred C57BL
- Cells, Cultured
- Signal Transduction
- Humans
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Vascular Remodeling
Collapse
Affiliation(s)
- Xinyue Song
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Ya Xu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Mengnan Li
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Xiaoyu Guan
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Huiyu Liu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Jingya Zhang
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Hanliang Sun
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Cui Ma
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Lixin Zhang
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xijuan Zhao
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xiaodong Zheng
- Department of Genetic and Cell Biology (X. Zheng), Harbin Medical University (Daqing), P.R. China
| | - Daling Zhu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education (D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| |
Collapse
|
2
|
Ochodnicka-Mackovicova K, van Keimpema M, Spaargaren M, van Noesel CJM, Guikema JEJ. DNA damage-induced p53 downregulates expression of RAG1 through a negative feedback loop involving miR-34a and FOXP1. J Biol Chem 2024; 300:107922. [PMID: 39454960 PMCID: PMC11625342 DOI: 10.1016/j.jbc.2024.107922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
During the maturation of pre-B cells, the recombination activating gene 1 and 2 (RAG1/2) endonuclease complex plays a crucial role in coordinating V(D)J recombination by introducing DNA breaks in immunoglobulin (Ig) loci. Dysregulation of RAG1/2 has been linked to the onset of B cell malignancies, yet the mechanisms controlling RAG1/2 in pre-B cells exposed to excessive DNA damage are not fully understood. In this study, we show that DNA damage-induced activation of p53 initiates a negative-feedback loop which rapidly downregulates RAG1 levels. This feedback loop involves ataxia telangiectasia mutated activation, subsequent stabilization of p53, and modulation of microRNA-34a (miR-34a) levels, which is one of the p53 targets. Notably, this loop incorporates transcription factor forkhead box P1 as a downstream effector. The absence of p53 resulted in an increased proportion of IgM+ cells prompted to upregulate RAG1/2 and to undergo Ig light chain recombination. Similar results were obtained in primary pre-B cells with depleted levels of miR-34a. We propose that in pre-B cells undergoing Ig gene recombination, the DNA breaks activate a p53/miR-34a/forkhead box P1-mediated negative-feedback loop that contributes to the rapid downregulation of RAG. This regulation limits the RAG-dependent DNA damage, thereby protecting the stability of the genome during V(D)J rearrangement in developing B cells.
Collapse
Affiliation(s)
- Katarina Ochodnicka-Mackovicova
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands
| | - Martine van Keimpema
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Carel J M van Noesel
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands
| | - Jeroen E J Guikema
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Dirimtekin E, Mortoglou M, Alavanda C, Benomar Yemlahi A, Arslan Ates E, Guney I, Uysal-Onganer P. miR-34a-FOXP1 Loop in Ovarian Cancer. ACS OMEGA 2023; 8:27743-27750. [PMID: 37546627 PMCID: PMC10399168 DOI: 10.1021/acsomega.3c03867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023]
Abstract
Ovarian cancer (OC) is the main cause of gynecological cancer mortality in most developed countries. microRNA (miR) expression dysregulation has been highlighted in human cancers, and miR-34a is found to be downregulated and associated with inhibition of tumor growth and invasion in several malignancies, including OC. The winged helix transcription factor forkhead box P1 (FOXP1) is reported as either an oncogene or tumor suppressor in various cancers. This study aimed to elucidate potential clinical and biological associations of miR-34a and transcription factor FOXP1 in OC. We investigated nine OC patients' blood samples and two OC cell lines (SKOV-3 and OVCAR-3) using quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR) to determine both miR-34a and FOXP1 expressions. We have found that miR-34a and FOXP1 are reversely correlated in both in vitro and in vivo. Inhibition of miR-34a transiently led to upregulation of FOXP1 mRNA expression and increased cellular invasion in vitro. Our data indicate that miR-34a could be a potential biomarker for improving the diagnostic efficiency of OC, and miR-34a overexpression may reduce OC pathogenesis by targeting FOXP1.
Collapse
Affiliation(s)
- Esra Dirimtekin
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
| | - Maria Mortoglou
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, U.K.
| | - Ceren Alavanda
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
- Department
of Medical Genetics, Van Training and Research Hospital, University of Health Sciences, 65170 Van, Turkey
| | - Asmaa Benomar Yemlahi
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, U.K.
| | - Esra Arslan Ates
- Department
of Medical Genetics, Istanbul University-Cerrahpasa,
Cerrahpasa Faculty of Medicine, 34098 Istanbul, Turkey
| | - Ilter Guney
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
| | - Pinar Uysal-Onganer
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, U.K.
| |
Collapse
|
4
|
Ismailova A, Salehi-Tabar R, Dimitrov V, Memari B, Barbier C, White JH. Identification of a forkhead box protein transcriptional network induced in human neutrophils in response to inflammatory stimuli. Front Immunol 2023; 14:1123344. [PMID: 36756115 PMCID: PMC9900176 DOI: 10.3389/fimmu.2023.1123344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
Introduction Neutrophils represent the largest proportion of circulating leukocytes and, in response to inflammatory stimuli, are rapidly recruited to sites of infection where they neutralize pathogens. Methods and results We have identified a novel neutrophil transcription network induced in response to inflammatory stimuli. We performed the first RNAseq analysis of human neutrophils exposed to lipopolysaccharide (LPS), followed by a meta-analysis of our dataset and previously published studies of LPS-challenged neutrophils. This revealed a robustly enhanced transcriptional network driven by forkhead box (FOX) transcription factors. The network is enriched in genes encoding proinflammatory cytokines and transcription factors, including MAFF and ATF3, which are implicated in responses to stress, survival and inflammation. Expression of transcription factors FOXP1 and FOXP4 is induced in neutrophils exposed to inflammatory stimuli, and potential FOXP1/FOXP4 binding sites were identified in several genes in the network, all located in chromatin regions consistent with neutrophil enhancer function. Chromatin immunoprecipitation (ChIP) assays in neutrophils confirmed enhanced binding of FOXP4, but not FOXP1, to multiple sites in response to LPS. Binding to numerous motifs and transactivation of network genes were also observed when FOXP proteins were transiently expressed in HEK293 cells. In addition to LPS, the transcriptional network is induced by other inflammatory stimuli, indicating it represents a general neutrophil response to inflammation. Discussion Collectively, these findings reveal a role for the FOXP4 transcription network as a regulator of responses to inflammatory stimuli in neutrophils.
Collapse
Affiliation(s)
- Aiten Ismailova
- Department of Physiology, McGill University, Montreal, QC, Canada
| | | | - Vassil Dimitrov
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Babak Memari
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Camille Barbier
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - John H. White
- Department of Physiology, McGill University, Montreal, QC, Canada,Department of Medicine, McGill University, Montreal, QC, Canada,*Correspondence: John H. White,
| |
Collapse
|
5
|
Hu YZ, Li Q, Wang PF, Li XP, Hu ZL. Multiple functions and regulatory network of miR-150 in B lymphocyte-related diseases. Front Oncol 2023; 13:1140813. [PMID: 37182123 PMCID: PMC10172652 DOI: 10.3389/fonc.2023.1140813] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
MicroRNAs (miRNAs) play vital roles in the post-transcriptional regulation of gene expression. Previous studies have shown that miR-150 is a crucial regulator of B cell proliferation, differentiation, metabolism, and apoptosis. miR-150 regulates the immune homeostasis during the development of obesity and is aberrantly expressed in multiple B-cell-related malignant tumors. Additionally, the altered expression of MIR-150 is a diagnostic biomarker of various autoimmune diseases. Furthermore, exosome-derived miR-150 is considered as prognostic tool in B cell lymphoma, autoimmune diseases and immune-mediated disorders, suggesting miR-150 plays a vital role in disease onset and progression. In this review, we summarized the miR-150-dependent regulation of B cell function in B cell-related immune diseases.
Collapse
Affiliation(s)
- Yue-Zi Hu
- Clinical Laboratory, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qiao Li
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Fei Wang
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Ping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhao-Lan Hu,
| |
Collapse
|
6
|
Kaminskiy Y, Kuznetsova V, Kudriaeva A, Zmievskaya E, Bulatov E. Neglected, yet significant role of FOXP1 in T-cell quiescence, differentiation and exhaustion. Front Immunol 2022; 13:971045. [PMID: 36268015 PMCID: PMC9576946 DOI: 10.3389/fimmu.2022.971045] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/20/2022] [Indexed: 12/04/2022] Open
Abstract
FOXP1 is ubiquitously expressed in the human body and is implicated in both physiological and pathological processes including cancer. However, despite its importance the role of FOXP1 in T-cells has not been extensively studied. Although relatively few phenotypic and mechanistic details are available, FOXP1 role in T-cell quiescence and differentiation of CD4+ subsets has recently been established. FOXP1 prevents spontaneous T-cell activation, preserves memory potential, and regulates the development of follicular helper and regulatory T-cells. Moreover, there is growing evidence that FOXP1 also regulates T-cell exhaustion. Altogether this makes FOXP1 a crucial and highly undervalued regulator of T-cell homeostasis. In this review, we discuss the biology of FOXP1 with a focus on discoveries made in T-cells in recent years.
Collapse
Affiliation(s)
- Yaroslav Kaminskiy
- Department of Oncology and Pathology, Karolinska Institutet, SciLifeLab, Solna, Sweden
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, Moscow, Russia
| | - Varvara Kuznetsova
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, Moscow, Russia
| | - Anna Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina Zmievskaya
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Emil Bulatov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- *Correspondence: Emil Bulatov,
| |
Collapse
|
7
|
Braden RO, Amor DJ, Fisher SE, Mei C, Myers CT, Mefford H, Gill D, Srivastava S, Swanson LC, Goel H, Scheffer IE, Morgan AT. Severe speech impairment is a distinguishing feature of FOXP1-related disorder. Dev Med Child Neurol 2021; 63:1417-1426. [PMID: 34109629 DOI: 10.1111/dmcn.14955] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 12/21/2022]
Abstract
AIM To delineate the speech and language phenotype of a cohort of individuals with FOXP1-related disorder. METHOD We administered a standardized test battery to examine speech and oral motor function, receptive and expressive language, non-verbal cognition, and adaptive behaviour. Clinical history and cognitive assessments were analysed together with speech and language findings. RESULTS Twenty-nine patients (17 females, 12 males; mean age 9y 6mo; median age 8y [range 2y 7mo-33y]; SD 6y 5mo) with pathogenic FOXP1 variants (14 truncating, three missense, three splice site, one in-frame deletion, eight cytogenic deletions; 28 out of 29 were de novo variants) were studied. All had atypical speech, with 21 being verbal and eight minimally verbal. All verbal patients had dysarthric and apraxic features, with phonological deficits in most (14 out of 16). Language scores were low overall. In the 21 individuals who carried truncating or splice site variants and small deletions, expressive abilities were relatively preserved compared with comprehension. INTERPRETATION FOXP1-related disorder is characterized by a complex speech and language phenotype with prominent dysarthria, broader motor planning and programming deficits, and linguistic-based phonological errors. Diagnosis of the speech phenotype associated with FOXP1-related dysfunction will inform early targeted therapy. What this paper adds Individuals with FOXP1-related disorder have a complex speech and language phenotype. Dysarthria, which impairs intelligibility, is the dominant feature of the speech profile. No participants were receiving speech therapy for dysarthria, but were good candidates for therapy Features of speech apraxia occur alongside persistent phonological errors. Language abilities are low overall; however, expressive language is a relative strength.
Collapse
Affiliation(s)
- Ruth O Braden
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Audiology and Speech Pathology and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - David J Amor
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Audiology and Speech Pathology and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,The Royal Children's Hospital, Parkville, VIC, Australia.,Victorian Clinical Genetics Service, Parkville, VIC, Australia
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Cristina Mei
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Orygen and Centre for Youth Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Candace T Myers
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Heather Mefford
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Deepak Gill
- TY Nelson Department of Neurology, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | | | - Lindsay C Swanson
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Himanshu Goel
- Hunter Genetics, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Ingrid E Scheffer
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Audiology and Speech Pathology and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,The Royal Children's Hospital, Parkville, VIC, Australia.,Austin Health, Heidelberg, Melbourne, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Angela T Morgan
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Audiology and Speech Pathology and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,The Royal Children's Hospital, Parkville, VIC, Australia.,Victorian Clinical Genetics Service, Parkville, VIC, Australia
| |
Collapse
|
8
|
FOXP1 drives osteosarcoma development by repressing P21 and RB transcription downstream of P53. Oncogene 2021; 40:2785-2802. [PMID: 33716296 DOI: 10.1038/s41388-021-01742-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 01/31/2023]
Abstract
Osteosarcoma has a poor prognosis, and the poor understanding of the genetic drivers of osteosarcoma hinders further improvement in therapeutic approaches. Transcription factor forkhead box P1 (FOXP1) is a crucial modulator in skeletal development and aging. Here, we determined the role and regulatory mechanisms of FOXP1 in osteosarcoma. Higher FOXP1 expression correlated with malignancy in both osteosarcoma cell lines and clinical biopsies. FOXP1 overexpression and knockdown in osteosarcoma cell lines revealed that FOXP1 promoted proliferation, tumor sphere formation, migration and invasion, and inhibited anoikis. Mechanistically, FOXP1 acted as a repressor of P21 and RB (retinoblastoma protein) transcription, and directly interacted with the tumor suppressor p53 to inhibit its activity. Extracellular signal-regulated kinase/c-Jun N-terminal kinase (ERK/JNK) signaling and c-JUN/c-FOS transcription factors were found to be upstream activators of FOXP1. Moreover, FOXP1 silencing via lentivirus or adeno-associated virus (AAV)-mediated delivery of shRNA suppressed osteosarcoma development and progression in cell-derived and patient-derived xenograft animal models. Taken together, we demonstrate that FOXP1, which is transactivated by ERK/JNK-c-JUN/c-FOS, drives osteosarcoma development by regulating the p53-P21/RB signaling cascade, suggesting that FOXP1 is a potential target for osteosarcoma therapy.
Collapse
|
9
|
Felce SL, Anderson AP, Maguire S, Gascoyne DM, Armstrong RN, Wong KK, Li D, Banham AH. CRISPR/Cas9-Mediated Foxp1 Silencing Restores Immune Surveillance in an Immunocompetent A20 Lymphoma Model. Front Oncol 2020; 10:448. [PMID: 32309216 PMCID: PMC7145990 DOI: 10.3389/fonc.2020.00448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/13/2020] [Indexed: 11/13/2022] Open
Abstract
The interaction of lymphoma cells with their microenvironment has an important role in disease pathogenesis and is being actively pursued therapeutically using immunomodulatory drugs, including immune checkpoint inhibitors. Diffuse large B-cell lymphoma (DLBCL) is an aggressive high-grade disease that remains incurable in ~40% of patients treated with R-CHOP immunochemotherapy. The FOXP1 transcription factor is abundantly expressed in such high-risk DLBCL and we recently identified its regulation of immune response signatures, in particular, its suppression of the cell surface expression of major histocompatibility class II (MHC-II), which has a critical role in antigen presentation to T cells. Using CRISPR/Cas9 genome editing we have depleted Foxp1 expression in the aggressive murine A20 lymphoma cell line. When grown in BALB/c mice, this cell line provides a high-fidelity immunocompetent disseminated lymphoma model that displays many characteristics of human DLBCL. Transient Foxp1-depletion using siRNA, and stable depletion using CRISPR (generated by independently targeting Foxp1 exon six or seven) upregulated cell surface I-Ab (MHC-II) expression without impairing cell viability in vitro. RNA sequencing of Foxp1-depleted A20 clones identified commonly deregulated genes, such as the B-cell marker Cd19, and hallmark DLBCL signatures such as MYC-targets and oxidative phosphorylation. Immunocompetent animals bearing Foxp1-depleted A20 lymphomas showed significantly-improved survival, and 20% did not develop tumors; consistent with modulating immune surveillance, this was not observed in immunodeficient NOD SCIDγ mice. The A20 Foxp1 CRISPR model will help to further characterize the contribution of Foxp1 to lymphoma immune evasion and the potential for Foxp1 targeting to synergize with other immunotherapies.
Collapse
Affiliation(s)
- Suet Ling Felce
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Amanda P. Anderson
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Shaun Maguire
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Duncan M. Gascoyne
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Richard N. Armstrong
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Genetics and Genome Biology Program, Haematology Section, Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- The Marrow Failure and Myelodysplasia Program, Haematology Section, Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Health, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Demin Li
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison H. Banham
- NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Wang Y, Zhang H, Jiao B, Nie J, Li X, Wang W, Wang H. The Roles of Alternative Splicing in Tumor-immune Cell Interactions. Anticancer Agents Med Chem 2020; 20:729-740. [PMID: 32560607 PMCID: PMC8388066 DOI: 10.2174/1568009620666200619123725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022]
Abstract
Alternative splicing (AS) plays a significant role in the hallmarks of cancer and can provide neoantigens for immunotherapy. Here, we summarize recent advances in immune system associated tumor specific-antigens (TSAs) produced by AS. We further discuss the regulating mechanisms involved in AS-mediated innate and adaptive immune responses and the anti-tumoral and protumoral roles in different types of cancer. For example, ULBP1_RI, MLL5Δ21spe, NKp44-1Δ5, MHC-IΔ7, CD200SΔ1, 2, PVR α/β/γ/δ and IL-33 variants 1/2/3 act as regulators in solid tumors and IPAK4-L and, FOXP1ΔN100 exhibit functions in hematological cancers.
Collapse
Affiliation(s)
| | - Honglei Zhang
- Address correspondence to these authors at Kunming Institute of Zoology, Chinese Academy of Sciences; 32 Jiaochang E. Road, Kunming, Yunnan, China; Tel: +86-871-68191706; E-mail: ; and Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan, China; Tel: +86-13608815577; E-mail:
| | - Baowei Jiao
- Address correspondence to these authors at Kunming Institute of Zoology, Chinese Academy of Sciences; 32 Jiaochang E. Road, Kunming, Yunnan, China; Tel: +86-871-68191706; E-mail: ; and Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan, China; Tel: +86-13608815577; E-mail:
| | - Jianyun Nie
- Address correspondence to these authors at Kunming Institute of Zoology, Chinese Academy of Sciences; 32 Jiaochang E. Road, Kunming, Yunnan, China; Tel: +86-871-68191706; E-mail: ; and Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan, China; Tel: +86-13608815577; E-mail:
| | | | | | | |
Collapse
|
11
|
Harb OA, Kaf RM, Taha HF, Ahmed RZ, Mandour D, Al Attar AZ, Fathy A, Almoregy AS, Osman G, Gertallah LM. Prognostic values and clinical implications of programmed cell death-ligand 1 (PD-L1), fork head transcription factor P-1 (FOXP-1) and signal transducer and activator of transcription-3 (STAT-3) expression in diffuse large B-cell lymphoma (DLBCL); an immunohistochemical study. SURGICAL AND EXPERIMENTAL PATHOLOGY 2019. [DOI: 10.1186/s42047-019-0038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Context
PD-L1 is an inhibitory ligand that functions as an essential immune checkpoint. FOXP-1 is a member of the FOXP family. STAT-3 plays a critical role in regulation of cell proliferation and survival. The detailed expression of the three markers together in DLBCL tissues and their prognostic value in patients with DLBCL were not fully investigated.
Aim was to assess the expression of PD-L1, FOXP-1 and STAT-3 in diffuse large B-cell lymphoma (DLBCL) and to correlate their expression with the pathological findings, prognostic parameters and clinical implications of patients.
Methods
PD-L1, FOXP-1 and STAT-3 were assessed in DLBCL tissues derived from 50 patients using immunohistochemistry. Patients were followed up for 3 years for response to therapy progression, recurrence and survival.
Results
High PD-L1 expression was associated with bone marrow involvement (p = 0.004), extra-nodal involvement (p = 0.006) and advanced stage (p = 0.003). High FOXP-1 expression was associated with presence of bone marrow involvement and high risk group (p < 0.001). High STAT-3 expression was associated with older age of the patient (p < 0.001), presence of bone marrow involvement (p = 0.002), extra-nodal involvement (p = 0.009), and high risk group (p = 0.005). High expression of PD-L1, FOXP-1 and STAT-3 was related to poor response to therapy, poor OS rate and RFS rates (p < 0.001).
Conclusion
High expression of PD-L1, FOXP-1 and STAT-3 was related poor prognosis in DLBCL patients.
Collapse
|
12
|
PD-1/PD-L1 immune checkpoint and p53 loss facilitate tumor progression in activated B-cell diffuse large B-cell lymphomas. Blood 2019; 133:2401-2412. [PMID: 30975638 DOI: 10.1182/blood.2018889931] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Refractory or relapsed diffuse large B-cell lymphoma (DLBCL) often associates with the activated B-cell-like (ABC) subtype and genetic alterations that drive constitutive NF-κB activation and impair B-cell terminal differentiation. Here, we show that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from Blimp1 loss in ABC-DLBCL lymphomagenesis. We provide evidences that the interplay between these genetic alterations and the tumor microenvironment select for additional molecular addictions that promote lymphoma progression, including aberrant coexpression of FOXP1 and the B-cell mutagenic enzyme activation-induced deaminase, and immune evasion through major histocompatibility complex class II downregulation, PD-L1 upregulation, and T-cell exhaustion. Consistently, PD-1 blockade cooperated with anti-CD20-mediated B-cell cytotoxicity, promoting extended T-cell reactivation and antitumor specificity that improved long-term overall survival in mice. Our data support a pathogenic cooperation among NF-κB-driven prosurvival, genetic instability, and immune evasion mechanisms in DLBCL and provide preclinical proof of concept for including PD-1/PD-L1 blockade in combinatorial immunotherapy for ABC-DLBCL.
Collapse
|
13
|
Jespersen DS, Schönherz AA, Due H, Bøgsted M, Sondergaard TE, Dybkær K. Expression of NOTCH3 exon 16 differentiates Diffuse Large B-cell Lymphoma into molecular subtypes and is associated with prognosis. Sci Rep 2019; 9:335. [PMID: 30674940 PMCID: PMC6344585 DOI: 10.1038/s41598-018-36680-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/23/2018] [Indexed: 01/19/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease with diverse clinical presentation and outcome. Bio-clinical prognostic models including oncogene expression and cell-of-origin phenotyping has been developed, however, approximately 30% of all patients still die from their disease, illustrating the need for additional prognostic biomarkers associating oncogenesis and phenotypic subclasses. Hence, we tested if alternative splice variations have biomarker potential. Initial alternative splicing analysis of human exon array from clinical DLBCL samples identified candidate genes. Experimental validation by ddPCR was performed in a DLBCL cohort classified into ABC/GCB subclasses, B-cell associated gene signatures (BAGS: naive, centroblast, centrocyte, memory, and plasmablast), and vincristine resistant gene signatures. Prognostic potential was assessed for aberrantly spliced transcripts. Thus, NOTCH3 was identified as alternatively spliced, with differential exon 16 depletion (−exon 16) between differentiation associated BAGS subtypes. Predicted vincristine resistant patients of the GCB subclass had significantly downregulated NOTCH3 −exon 16 transcript expression and tended to display adverse overall survival for R-CHOP treated patients. In conclusion, we have identified a specific alternatively spliced NOTCH3 event that differentiate molecular subtypes of DLBCL and display prognostic and predictive biomarker potential in GCB DLBCL.
Collapse
Affiliation(s)
| | - Anna A Schönherz
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark. .,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark. .,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
14
|
miR-150 downregulation contributes to the high-grade transformation of follicular lymphoma by upregulating FOXP1 levels. Blood 2018; 132:2389-2400. [PMID: 30213873 DOI: 10.1182/blood-2018-06-855502] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 09/05/2018] [Indexed: 12/12/2022] Open
Abstract
Follicular lymphoma (FL) is a common indolent B-cell malignancy with a variable clinical course. An unfavorable event in its course is histological transformation to a high-grade lymphoma, typically diffuse large B-cell lymphoma. Recent studies show that genetic aberrations of MYC or its overexpression are associated with FL transformation (tFL). However, the precise molecular mechanisms underlying tFL are unclear. Here we performed the first profiling of expression of microRNAs (miRNAs) in paired samples of FL and tFL and identified 5 miRNAs as being differentially expressed. We focused on one of these miRNAs, namely miR-150, which was uniformly downmodulated in all examined tFLs (∼3.5-fold), and observed that high levels of MYC are responsible for repressing miR-150 in tFL by binding in its upstream region. This MYC-mediated repression of miR-150 in B cells is not dependent on LIN28A/B proteins, which influence the maturation of miR-150 precursor (pri-miR-150) in myeloid cells. We also demonstrated that low miR-150 levels in tFL lead to upregulation of its target, namely FOXP1 protein, which is a known positive regulator of cell survival, as well as B-cell receptor and NF-κB signaling in malignant B cells. We revealed that low levels of miR-150 and high levels of its target, FOXP1, are associated with shorter overall survival in FL and suggest that miR-150 could serve as a good biomarker measurable in formalin-fixed paraffin-embedded tissue. Overall, our study demonstrates the role of the MYC/miR-150/FOXP1 axis in malignant B cells as a determinant of FL aggressiveness and its high-grade transformation.
Collapse
|
15
|
MicroRNA miR-34a downregulates FOXP1 during DNA damage response to limit BCR signalling in chronic lymphocytic leukaemia B cells. Leukemia 2018; 33:403-414. [PMID: 30111844 DOI: 10.1038/s41375-018-0230-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/03/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022]
Abstract
The variable clinical course in chronic lymphocytic leukaemia (CLL) largely depends on p53 functionality and B-cell receptor (BCR) signalling propensity; however, it is unclear if there is any crosstalk between these pathways. We show that DNA damage response (DDR) activation leads to down-modulating the transcriptional factor FOXP1, which functions as a positive BCR signalling regulator and its high levels are associated with worse CLL prognosis. We identified microRNA (miRNA) miR-34a as the most prominently upregulated miRNA during DDR in CLL cells in vitro and in vivo during FCR therapy (fludarabine, cyclophosphamide, rituximab). MiR-34a induced by DDR activation and p53 stabilization potently represses FOXP1 expression by binding in its 3'-UTR. The low FOXP1 levels limit BCR signalling partially via derepressing BCR-inhibitory molecule CD22. We also show that low miR-34a levels can be used as a biomarker for worse response or shorter progression free survival in CLL patients treated with FCR chemoimmunotherapy, and shorter overall survival, irrespective of TP53 status. Additionally, we have developed a method for the absolute quantification of miR-34a copies and defined precise prognostic/predictive cutoffs. Overall, herein, we reveal for the first time that B cells limit their BCR signalling during DDR by down-modulating FOXP1 via DDR-p53/miR-34a axis.
Collapse
|