1
|
Levavasseur F, Oussous S, Framarini A, Boussaid I, Gou P, Tuerdi Z, Boueya IL, Hoffner H, De Almeida M, Gall ML, Tucker H, Giraudier S, Bouscary D, Fontenay M, Passaro D, Dusanter-Fourt I, Lauret E. FOXP1 contributes to murine hematopoietic stem cell functionality. Exp Hematol 2025:104815. [PMID: 40449872 DOI: 10.1016/j.exphem.2025.104815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/23/2025] [Accepted: 05/13/2025] [Indexed: 06/03/2025]
Abstract
Transcription factor forkhead box P1 (FOXP1) is a key regulator of immune cell functions. We have shown that FOXP1 contributes to the expansion of human hematopoietic stem/progenitor (HSPC) and acute myeloid leukaemia cells. Here, we investigated the role of FOXP1 in early adult mouse hematopoiesis in vivo. We showed that loss of hematopoietic-specific FOXP1 expression leads to attrition of the HSC and multipotent progenitor (MPP)-1 compartment in parallel with enhancement of myeloid-biased MPP3 in adult bone marrow and fetal liver. Transplantation experiments confirmed that FOXP1-deficient bone marrow had an intrinsic reduced HSC compartment. FOXP1-deficient MPP compartments also showed enhanced proliferation with G0 phase reduction. Transcriptome analyses revealed that FOXP1-deficient HSC exhibited reduced stemness and enhanced expression of cell proliferation pathways. Thus, our current results reveal the important contribution of FOXP1 in early murine hematopoiesis through HSC maintenance, limited expansion of all MPP compartments and restriction of early myeloid commitment in vivo.
Collapse
Affiliation(s)
- Françoise Levavasseur
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Samia Oussous
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Alessandro Framarini
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Ismael Boussaid
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France; Assistance Publique-Hôpitaux de Paris. Centre-Université Paris Cité, Hôpital Cochin, Laboratory of Hematology, Paris, France
| | - Panhong Gou
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France. Université de Paris Cité, Paris, France. Service de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Zubaidan Tuerdi
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Iman Litchy Boueya
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - Helyette Hoffner
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Marta De Almeida
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Morgane Le Gall
- Plateforme Proteom'IC, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Haley Tucker
- Departments of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Stéphane Giraudier
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France. Université de Paris Cité, Paris, France. Service de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Didier Bouscary
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France; Assistance Publique-Hôpitaux de Paris. Centre-Université Paris Cité, Hôpital Cochin, Laboratory of Hematology, Paris, France
| | - Michaela Fontenay
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France; Assistance Publique-Hôpitaux de Paris. Centre-Université Paris Cité, Hôpital Cochin, Laboratory of Hematology, Paris, France
| | - Diana Passaro
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - Isabelle Dusanter-Fourt
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France
| | - Evelyne Lauret
- Normal and Pathological Hematopoiesis laboratory, Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014 Paris, France.
| |
Collapse
|
2
|
Abraham E, Kostina A, Volmert B, Roule T, Huang L, Yu J, Williams AE, Megill E, Douglas A, Pericak OM, Morris A, Stronati E, Larrinaga-Zamanillo A, Fueyo R, Zubillaga M, Andrake MD, Akizu N, Aguirre A, Estaras C. A retinoic acid:YAP1 signaling axis controls atrial lineage commitment. Cell Rep 2025; 44:115687. [PMID: 40343798 DOI: 10.1016/j.celrep.2025.115687] [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: 08/23/2024] [Revised: 03/10/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
In cardiac progenitor cells (CPCs), retinoic acid (RA) signaling induces atrial lineage gene expression and acquisition of an atrial cell fate. To achieve this, RA coordinates a complex regulatory network of downstream effectors that is not fully identified. To address this gap, we applied a functional genomics approach (i.e., scRNA-seq and snATAC-seq) to untreated and RA-treated human embryonic stem cell (hESC)-derived CPCs. Unbiased analysis revealed that the Hippo effectors YAP1 and TEAD4 are integrated with the atrial transcription factor enhancer network and that YAP1 activates RA enhancers in CPCs. Furthermore, Yap1 deletion in mouse embryos compromises the expression of RA-induced genes, such as Nr2f2, in the CPCs of the second heart field. Accordingly, in hESC-derived patterned heart organoids, YAP1 regulates the formation of an atrial chamber but is dispensable for the formation of a ventricle. Overall, our findings revealed that YAP1 cooperates with RA signaling to induce atrial lineages during cardiogenesis.
Collapse
Affiliation(s)
- Elizabeth Abraham
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Aleksandra Kostina
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Brett Volmert
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Thomas Roule
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ling Huang
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jingting Yu
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - April E Williams
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emily Megill
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Aidan Douglas
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Olivia M Pericak
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Alex Morris
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Eleonora Stronati
- Department of Child and Adolescence Psychiatry, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Arantza Larrinaga-Zamanillo
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Raquel Fueyo
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mikel Zubillaga
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Mark D Andrake
- Molecular Modeling Facility, Program in Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Naiara Akizu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Conchi Estaras
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
3
|
Zhu X, Gao C, Peng B, Xue J, Xia D, Yang L, Zhang J, Gao X, Hu Y, Lin S, Gong P, Xu X. FOXP1 phosphorylation antagonizes its O-GlcNAcylation in regulating ATR activation in response to replication stress. EMBO J 2025; 44:457-483. [PMID: 39623140 PMCID: PMC11729909 DOI: 10.1038/s44318-024-00323-x] [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: 04/18/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 01/15/2025] Open
Abstract
ATR signaling is essential in sensing and responding to the replication stress; as such, any defects can impair cellular function and survival. ATR itself is activated via tightly regulated mechanisms. Here, we identify FOXP1, a forkhead-box-containing transcription factor, as a regulator coordinating ATR activation. We show that, unlike its role as a transcription factor, FOXP1 functions as a scaffold and directly binds to RPA-ssDNA and ATR-ATRIP complexes, facilitating the recruitment and activation of ATR. This process is regulated by FOXP1 O-GlcNAcylation, which represses its interaction with ATR, while CHK1-mediated phosphorylation of FOXP1 inhibits its O-GlcNAcylation upon replication stress. Supporting the physiological relevance of this loop, we find pathogenic FOXP1 mutants identified in various tumor tissues with compromised ATR activation and stalled replication fork stability. We thus conclude that FOXP1 may serve as a potential chemotherapeutic target in related tumors.
Collapse
Affiliation(s)
- Xuefei Zhu
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| | - Congwen Gao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- College of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, 071002, Baoding, China
| | - Bin Peng
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Jingwei Xue
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Donghui Xia
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Shenzhen University General Hospital-Dehua Hospital Joint Research Center on Precision Medicine (sgh-dhhCPM), Dehua Hospital, Dehua, 362500, Quanzhou, China
- State Key Laboratory of Agro-biotechnology and MOA Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, 100193, Beijing, China
| | - Liu Yang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Jiexiang Zhang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, 518060, Shenzhen, Guangdong, China
| | - Xinrui Gao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Yilin Hu
- Life Sciences Institute, Zhejiang University, 310058, Hangzhou, China
| | - Shixian Lin
- Life Sciences Institute, Zhejiang University, 310058, Hangzhou, China
| | - Peng Gong
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| | - Xingzhi Xu
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Schonk MM, Ducharme JB, Neyroud D, Nosacka RL, Tucker HO, Judge SM, Judge AR. Role of myofiber-specific FoxP1 in pancreatic cancer-induced muscle wasting. Am J Physiol Cell Physiol 2025; 328:C1-C8. [PMID: 39545377 DOI: 10.1152/ajpcell.00701.2024] [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: 09/23/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Cancer cachexia affects up to 80% of patients with cancer and results in reduced quality of life and survival. We previously demonstrated that the transcriptional repressor Forkhead box P1 (FoxP1) is upregulated in the skeletal muscle of cachectic mice and people with cancer, and when overexpressed in skeletal muscle, it is sufficient to induce pathological features characteristic of cachexia. However, the role of myofiber-derived FoxP1 in both normal muscle physiology and cancer-induced muscle wasting remains largely unexplored. To address this gap, we generated a conditional mouse line with myofiber-specific ablation of FoxP1 (FoxP1SkmKO) and found that in cancer-free mice, deletion of FoxP1 in skeletal myofibers resulted in increased myofiber size in both males and females, with a significant increase in muscle mass in males. In response to murine KPC pancreatic tumor burden, we found that myofiber-derived FoxP1 mediates cancer-induced muscle wasting and diaphragm muscle weakness in male but not female mice. In summary, our findings identify myofiber-specific FoxP1 as a negative regulator of skeletal muscle with sex-specific differences in the context of cancer.NEW & NOTEWORTHY Here we identify myofiber-derived FoxP1 as a negative regulator of skeletal muscle with sex-specific effects in cancer. Under cancer-free conditions, FoxP1 knockout increased myofiber size in male and female mice. However, in response to pancreatic cancer, FoxP1 myofiber-specific deletion attenuated muscle wasting and weakness in males but not females. This highlights the need to consider sexual dimorphism in cancer-induced muscle pathologies and provides evidence suggesting that targeting FoxP1 could help mitigate these effects in males.
Collapse
Affiliation(s)
- Martin M Schonk
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Jeremy B Ducharme
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
| | - Daria Neyroud
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Rachel L Nosacka
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
| | - Haley O Tucker
- Department of Molecular Biosciences and the Institute for Cellular and Molecular Biology, University of Texas at Austin, Texas, United States
| | - Sarah M Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Andrew R Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States
- Myology Institute, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
5
|
Hasan MR, Kump AJ, Stepaniak EC, Panta M, Shashidhar K, Katariya R, Sabbir MK, Schwab KR, Inlow MH, Chen Y, Ahmad SM. Genome-Wide Expression Profiling and Phenotypic Analysis of Downstream Targets Identify the Fox Transcription Factor Jumeau as a Master Regulator of Cardiac Progenitor Cell Division. Int J Mol Sci 2024; 25:12933. [PMID: 39684645 DOI: 10.3390/ijms252312933] [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: 09/16/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Forkhead box (Fox) transcription factors (TFs) mediate multiple conserved cardiogenic processes in both mammals and Drosophila. Our prior work identified the roles of two Drosophila Fox genes, jumeau (jumu) and Checkpoint suppressor 1-like (CHES-1-like), in cardiac progenitor cell specification and division, and in the proper positioning of cardiac cell subtypes. Fox TF binding sites are also significantly enriched in the enhancers of genes expressed in the heart, suggesting that these genes may play a core regulatory role in one or more of these cardiogenic processes. We identified downstream targets of Jumu by comparing transcriptional expression profiles of flow cytometry-sorted mesodermal cells from wild-type embryos and embryos completely lacking the jumu gene and found that genes with functional annotation and ontological features suggesting roles in cell division were overrepresented among Jumu targets. Phenotypic analysis of a subset of these targets identified 21 jumu-regulated genes that mediate cardiac progenitor cell division, one of which, Retinal Homeobox (Rx), was characterized in more detail. Finally, the observation that many of these 21 genes and/or their orthologs exhibit genetic or physical interactions among themselves indicates that Jumu is a master regulator acting as a hub of a cardiac progenitor cell division-mediating network.
Collapse
Affiliation(s)
- M Rezaul Hasan
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Andrew J Kump
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Evelyn C Stepaniak
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manoj Panta
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kuncha Shashidhar
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Rajnandani Katariya
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mofazzal K Sabbir
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kristopher R Schwab
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mark H Inlow
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Department of Mathematical Sciences, Indiana State University, Terre Haute, IN 47809, USA
| | - Ye Chen
- Department of Mathematics and Statistics, Northern Arizona University, Flagstaff, AZ 86001, USA
| | - Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| |
Collapse
|
6
|
Schonk MM, Ducharme JB, Neyroud D, Nosacka RL, Tucker HO, Judge SM, Judge AR. Myofiber-specific FoxP1 knockout protects against pancreatic cancer-induced muscle wasting in male but not female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613547. [PMID: 39345535 PMCID: PMC11429864 DOI: 10.1101/2024.09.17.613547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cancer cachexia affects up to 80% of cancer patients and results in reduced quality of life and survival. We previously demonstrated that the transcriptional repressor Forkhead box P1 (FoxP1) is upregulated in skeletal muscle of cachectic mice and people with cancer, and when overexpressed in skeletal muscle is sufficient to induce pathological features characteristic of cachexia. However, the role of myofiber-derived FoxP1 in both normal muscle physiology and cancer-induced muscle wasting remains largely unexplored. To address this gap, we generated a conditional mouse line with myofiber-specific ablation of FoxP1 (FoxP1SkmKO) and found that in cancer-free mice, deletion of FoxP1 in skeletal myofibers resulted in increased myofiber size in both males and females, with a significant increase in muscle mass in males. In response to murine KPC pancreatic tumor burden, we found that myofiber-derived FoxP1 is required for cancer-induced muscle wasting and diaphragm muscle weakness in male mice. In summary, our findings identify myofiber-specific FoxP1 as a negative regulator of skeletal muscle with sex-specific differences in the context of cancer.
Collapse
Affiliation(s)
- Martin M Schonk
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jeremy B Ducharme
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Daria Neyroud
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel L Nosacka
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Haley O Tucker
- Department of Molecular Biosciences and the Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Sarah M Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Andrew R Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Zsigmond A, Till Á, Bene J, Czakó M, Mikó A, Hadzsiev K. Case Report of Suspected Gonadal Mosaicism in FOXP1-Related Neurodevelopmental Disorder. Int J Mol Sci 2024; 25:5709. [PMID: 38891897 PMCID: PMC11171548 DOI: 10.3390/ijms25115709] [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: 04/27/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Heterozygous mutations in the FOXP1 gene (OMIM#605515) are responsible for a well-characterized neurodevelopmental syndrome known as "intellectual developmental disorder with language impairment with or without autistic features" (OMIM#613670) or FOXP1 syndrome for short. The main features of the condition are global developmental delay/intellectual disability; speech impairment in all individuals, regardless of their level of cognitive abilities; behavioral abnormalities; congenital anomalies, including subtle dysmorphic features; and strabismus, brain, cardiac, and urogenital abnormalities. Here, we present two siblings with a de novo heterozygous FOXP1 variant, namely, a four-year-old boy and 14-month-old girl. Both children have significantly delayed early psychomotor development, hypotonia, and very similar, slightly dysmorphic facial features. A lack of expressive speech was the leading symptom in the case of the four-year-old boy. We performed whole-exome sequencing on the male patient, which identified a pathogenic heterozygous c.1541G>A (p.Arg514His) FOXP1 mutation. His sister's targeted mutation analysis also showed the same heterozygous FOXP1 variant. Segregation analysis revealed the de novo origin of the mutation, suggesting the presence of parental gonadal mosaicism. To the best of our knowledge, this is the first report of gonadal mosaicism in FOXP1-related neurodevelopmental disorders in the medical literature.
Collapse
Affiliation(s)
- Anna Zsigmond
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
| | - Ágnes Till
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
| | - Judit Bene
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
| | - Márta Czakó
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
| | - Alexandra Mikó
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
- Institute for Translational Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Kinga Hadzsiev
- Department of Medical Genetics, Medical School, University of Pécs, H-7623 Pécs, Hungary; (A.Z.); (Á.T.); (J.B.); (M.C.); (A.M.)
| |
Collapse
|
8
|
Wu M, Tang W, Chen Y, Xue L, Dai J, Li Y, Zhu X, Wu C, Xiong J, Zhang J, Wu T, Zhou S, Chen D, Sun C, Yu J, Li H, Guo Y, Huang Y, Zhu Q, Wei S, Zhou Z, Wu M, Li Y, Xiang T, Qiao H, Wang S. Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1. NATURE AGING 2024; 4:527-545. [PMID: 38594460 PMCID: PMC11031396 DOI: 10.1038/s43587-024-00607-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/05/2024] [Indexed: 04/11/2024]
Abstract
Limited understanding exists regarding how aging impacts the cellular and molecular aspects of the human ovary. This study combines single-cell RNA sequencing and spatial transcriptomics to systematically characterize human ovarian aging. Spatiotemporal molecular signatures of the eight types of ovarian cells during aging are observed. An analysis of age-associated changes in gene expression reveals that DNA damage response may be a key biological pathway in oocyte aging. Three granulosa cells subtypes and five theca and stromal cells subtypes, as well as their spatiotemporal transcriptomics changes during aging, are identified. FOXP1 emerges as a regulator of ovarian aging, declining with age and inhibiting CDKN1A transcription. Silencing FOXP1 results in premature ovarian insufficiency in mice. These findings offer a comprehensive understanding of spatiotemporal variability in human ovarian aging, aiding the prioritization of potential diagnostic biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jing Yu
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Sino-French Cooperative Central Lab, Shanghai Pudong Gongli Hospital, Secondary Military Medical University, Shanghai, China
| | - Hongyi Li
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Yibao Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Qingqing Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ziliang Zhou
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ya Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Tao Xiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | | | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| |
Collapse
|
9
|
Koene S, Ropers FG, Wieland J, Rybak T, Wildschut F, Berghuis D, Morgan A, Trelles MP, Scheepe JR, Bökenkamp R, Peeters-Scholte CMPCD, Braden R, Santen GWE. Clinical phenotype of FOXP1 syndrome: parent-reported medical signs and symptoms in 40 individuals. J Med Genet 2024; 61:399-404. [PMID: 38123995 DOI: 10.1136/jmg-2023-109537] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND The first studies on patients with forkhead-box protein P1 (FOXP1) syndrome reported associated global neurodevelopmental delay, autism symptomatology, dysmorphic features and cardiac and urogenital malformations. The aim of this study was to assess the prevalence of congenital abnormalities in an unbiased cohort of patients with FOXP1 syndrome and to document rare complications. METHODS Patients with FOXP1 syndrome were included, mostly diagnosed via whole-exome sequencing for neurodevelopmental delay. A parent-report questionnaire was used to assess medical signs and symptoms, including questions about features rated as most burdensome by patients and their family. RESULTS Forty individuals were included, 20 females and 20 males. The mean age at assessment was 13.2 years (median 8.5 years; range 2-54 years; ≥18 years n = 7). Seven adults were included. All patients had developmental problems, including cognitive, communication, social-emotional and motor delays. The most prevalent medical signs and symptoms include delayed bladder control, sleeping problems, hypermetropia, strabismus, sacral dimple, undescended testes, abnormal muscle tone and airway infections. The most burdensome complaints for patients with FOXP1 syndrome, as perceived by parents, include intellectual disability, impaired communication, behaviour problems, lack of age-appropriate self-reliance, attention problems and anxiety. According to parents, patients have quite similar reported symptoms, although incontinence, obsessions and a complex sensory profile have a higher ranking. CONCLUSION The results of this study may be used to further guide medical management and identify patient priorities for future research targeted on those features of FOXP1 syndrome that most impair quality of life of patients and their families.
Collapse
Affiliation(s)
- Saskia Koene
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Jannelien Wieland
- Department of Child and Adolescent Psychiatry, Leiden University Medical Center, Leiden, Netherlands
| | - Tamara Rybak
- 's Heeren Loo Zorggroep, Amersfoort, Netherlands
| | - Floor Wildschut
- Clinical Neurodevelopmental Sciences, Leiden University Clinical and Adolescent Child Studies, Leiden, Netherlands
| | - Dagmar Berghuis
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Angela Morgan
- Victorian Clinical Genetics Service and Speech and Language, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Maria Pilar Trelles
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry and Human Behaviour, Brown University, Providence, Rhode Island, USA
| | | | - Regina Bökenkamp
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ruth Braden
- Speech and Language, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
10
|
Steiman S, Miyake T, McDermott JC. FoxP1 Represses MEF2A in Striated Muscle. Mol Cell Biol 2024; 44:57-71. [PMID: 38483114 PMCID: PMC10950271 DOI: 10.1080/10985549.2024.2323959] [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: 09/06/2023] [Revised: 12/14/2023] [Accepted: 01/23/2024] [Indexed: 03/19/2024] Open
Abstract
Myocyte enhancer factor 2 (MEF2) proteins are involved in multiple developmental, physiological, and pathological processes in vertebrates. Protein-protein interactions underlie the plethora of biological processes impacted by MEF2A, necessitating a detailed characterization of the MEF2A interactome. A nanobody based affinity-purification/mass spectrometry strategy was employed to achieve this goal. Specifically, the MEF2A protein complexes were captured from myogenic lysates using a GFP-tagged MEF2A protein immobilized with a GBP-nanobody followed by LC-MS/MS proteomic analysis to identify MEF2A interactors. After bioinformatic analysis, we further characterized the interaction of MEF2A with a transcriptional repressor, FOXP1. FOXP1 coprecipitated with MEF2A in proliferating myogenic cells which diminished upon differentiation (myotube formation). Ectopic expression of FOXP1 inhibited MEF2A driven myogenic reporter genes (derived from the creatine kinase muscle and myogenin genes) and delayed induction of endogenous myogenin during differentiation. Conversely, FOXP1 depletion enhanced MEF2A transactivation properties and myogenin expression. The FoxP1:MEF2A interaction is also preserved in cardiomyocytes and FoxP1 depletion enhanced cardiomyocyte hypertrophy. FOXP1 prevented MEF2A phosphorylation and activation by the p38MAPK pathway. Overall, these data implicate FOXP1 in restricting MEF2A function in order to avoid premature differentiation in myogenic progenitors and also to possibly prevent re-activation of embryonic gene expression in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Sydney Steiman
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - John C. McDermott
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| |
Collapse
|
11
|
Li S, Yan RG, Gao X, He Z, Wu SX, Wang YJ, Zhang YW, Tao HP, Zhang XN, Jia GX, Yang QE. Single-cell transcriptome analyses reveal critical regulators of spermatogonial stem cell fate transitions. BMC Genomics 2024; 25:138. [PMID: 38310206 PMCID: PMC10837949 DOI: 10.1186/s12864-024-10072-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/31/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Spermatogonial stem cells (SSCs) are the foundation cells for continual spermatogenesis and germline regeneration in mammals. SSC activities reside in the undifferentiated spermatogonial population, and currently, the molecular identities of SSCs and their committed progenitors remain unclear. RESULTS We performed single-cell transcriptome analysis on isolated undifferentiated spermatogonia from mice to decipher the molecular signatures of SSC fate transitions. Through comprehensive analysis, we delineated the developmental trajectory and identified candidate transcription factors (TFs) involved in the fate transitions of SSCs and their progenitors in distinct states. Specifically, we characterized the Asingle spermatogonial subtype marked by the expression of Eomes. Eomes+ cells contained enriched transplantable SSCs, and more than 90% of the cells remained in the quiescent state. Conditional deletion of Eomes in the germline did not impact steady-state spermatogenesis but enhanced SSC regeneration. Forced expression of Eomes in spermatogenic cells disrupted spermatogenesis mainly by affecting the cell cycle progression of undifferentiated spermatogonia. After injury, Eomes+ cells re-enter the cell cycle and divide to expand the SSC pool. Eomes+ cells consisted of 7 different subsets of cells at single-cell resolution, and genes enriched in glycolysis/gluconeogenesis and the PI3/Akt signaling pathway participated in the SSC regeneration process. CONCLUSIONS In this study, we explored the molecular characteristics and critical regulators of subpopulations of undifferentiated spermatogonia. The findings of the present study described a quiescent SSC subpopulation, Eomes+ spermatogonia, and provided a dynamic transcriptional map of SSC fate determination.
Collapse
Affiliation(s)
- Shuang Li
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Medical Technology, Luoyang Polytechnic, Luoyang, Henan, 471000, China
| | - Rong-Ge Yan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue Gao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen He
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Xin Wu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu-Jun Wang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Wen Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hai-Ping Tao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Na Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Gong-Xue Jia
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810001, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810008, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, 810001, China.
| |
Collapse
|
12
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
13
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Semilunar Valve and Aortic Arch Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:777-796. [PMID: 38884748 DOI: 10.1007/978-3-031-44087-8_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The great arteries of the vertebrate carry blood from the heart to the systemic circulation and are derived from the pharyngeal arch arteries. In higher vertebrates, the pharyngeal arch arteries are a symmetrical series of blood vessels that rapidly remodel during development to become the asymmetric aortic arch arteries carrying oxygenated blood from the left ventricle via the outflow tract. At the base of the aorta, as well as the pulmonary trunk, are the semilunar valves. These valves each have three leaflets and prevent the backflow of blood into the heart. During development, the process of aortic arch and valve formation may go wrong, resulting in cardiovascular defects, and these may, at least in part, be caused by genetic mutations. In this chapter, we will review models harboring genetic mutations that result in cardiovascular defects affecting the great arteries and the semilunar valves.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK
| | - Simon D Bamforth
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK.
| |
Collapse
|
14
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Truncus Arteriosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:853-865. [PMID: 38884754 DOI: 10.1007/978-3-031-44087-8_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
In normal cardiovascular development in birds and mammals, the outflow tract of the heart is divided into two distinct channels to separate the oxygenated systemic blood flow from the deoxygenated pulmonary circulation. When the process of outflow tract septation fails, a single common outflow vessel persists resulting in a serious clinical condition known as persistent truncus arteriosus or common arterial trunk. In this chapter, we will review molecular pathways and the cells that are known to play a role in the formation and development of the outflow tract and how genetic manipulation of these pathways in animal models can result in common arterial trunk.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
15
|
Benvenuto M, Palumbo P, Di Muro E, Perrotta CS, Mazza T, Mandarà GML, Palumbo O, Carella M. Identification of a Novel FOXP1 Variant in a Patient with Hypotonia, Intellectual Disability, and Severe Speech Impairment. Genes (Basel) 2023; 14:1958. [PMID: 37895307 PMCID: PMC10606110 DOI: 10.3390/genes14101958] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
The FOXP subfamily includes four different transcription factors: FOXP1, FOXP2, FOXP3, and FOXP4, all with important roles in regulating gene expression from early development through adulthood. Haploinsufficiency of FOXP1, due to deleterious variants (point mutations, copy number variants) disrupting the gene, leads to an emerging disorder known as "FOXP1 syndrome", mainly characterized by intellectual disability, language impairment, dysmorphic features, and multiple congenital abnormalities with or without autistic features in some affected individuals (MIM 613670). Here we describe a 10-year-old female patient, born to unrelated parents, showing hypotonia, intellectual disability, and severe language delay. Targeted resequencing analysis allowed us to identify a heterozygous de novo FOXP1 variant c.1030C>T, p.(Gln344Ter) classified as likely pathogenetic according to the American College of Medical Genetics and Genomics guidelines. To the best of our knowledge, our patient is the first to date to report carrying this stop mutation, which is, for this reason, useful for broadening the molecular spectrum of FOXP1 clinically relevant variants. In addition, our results highlight the utility of next-generation sequencing in establishing an etiological basis for heterogeneous conditions such as neurodevelopmental disorders and providing additional insight into the phenotypic features of FOXP1-related syndrome.
Collapse
Affiliation(s)
- Mario Benvenuto
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.B.); (P.P.); (E.D.M.); (O.P.)
- Dipartimento Degli Studi Umanistici, Università Degli Studi di Foggia, 71122 Foggia, Italy
| | - Pietro Palumbo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.B.); (P.P.); (E.D.M.); (O.P.)
| | - Ester Di Muro
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.B.); (P.P.); (E.D.M.); (O.P.)
| | | | - Tommaso Mazza
- Unit of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | | | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.B.); (P.P.); (E.D.M.); (O.P.)
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.B.); (P.P.); (E.D.M.); (O.P.)
| |
Collapse
|
16
|
Li X, Hao S, Zou S, Tu X, Kong W, Jiang T, Chen JG. Cortex-restricted deletion of Foxp1 impairs barrel formation and induces aberrant tactile responses in a mouse model of autism. Mol Autism 2023; 14:34. [PMID: 37691105 PMCID: PMC10494400 DOI: 10.1186/s13229-023-00567-0] [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: 04/12/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Many children and young people with autism spectrum disorder (ASD) display touch defensiveness or avoidance (hypersensitivity), or engage in sensory seeking by touching people or objects (hyposensitivity). Abnormal sensory responses have also been noticed in mice lacking ASD-associated genes. Tactile sensory information is normally processed by the somatosensory system that travels along the thalamus to the primary somatosensory cortex. The neurobiology behind tactile sensory abnormalities, however, is not fully understood. METHODS We employed cortex-specific Foxp1 knockout (Foxp1-cKO) mice as a model of autism in this study. Tactile sensory deficits were measured by the adhesive removal test. The mice's behavior and neural activity were further evaluated by the whisker nuisance test and c-Fos immunofluorescence, respectively. We also studied the dendritic spines and barrel formation in the primary somatosensory cortex by Golgi staining and immunofluorescence. RESULTS Foxp1-cKO mice had a deferred response to the tactile environment. However, the mice exhibited avoidance behavior and hyper-reaction following repeated whisker stimulation, similar to a fight-or-flight response. In contrast to the wild-type, c-Fos was activated in the basolateral amygdala but not in layer IV of the primary somatosensory cortex of the cKO mice. Moreover, Foxp1 deficiency in cortical neurons altered the dendrite development, reduced the number of dendritic spines, and disrupted barrel formation in the somatosensory cortex, suggesting impaired somatosensory processing may underlie the aberrant tactile responses. LIMITATIONS It is still unclear how the defective thalamocortical connection gives rise to the hyper-reactive response. Future experiments with electrophysiological recording are needed to analyze the role of thalamo-cortical-amygdala circuits in the disinhibiting amygdala and enhanced fearful responses in the mouse model of autism. CONCLUSIONS Foxp1-cKO mice have tactile sensory deficits while exhibit hyper-reactivity, which may represent fearful and emotional responses controlled by the amygdala. This study presents anatomical evidence for reduced thalamocortical connectivity in a genetic mouse model of ASD and demonstrates that the cerebral cortex can be the origin of atypical sensory behaviors.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Shishuai Hao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Shimin Zou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Xiaomeng Tu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Weixi Kong
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Tian Jiang
- Research Center for Translational Medicine, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, 317500, People's Republic of China
| | - Jie-Guang Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, Zhejiang, People's Republic of China.
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| |
Collapse
|
17
|
Park SHE, Kulkarni A, Konopka G. FOXP1 orchestrates neurogenesis in human cortical basal radial glial cells. PLoS Biol 2023; 21:e3001852. [PMID: 37540706 PMCID: PMC10431666 DOI: 10.1371/journal.pbio.3001852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 08/16/2023] [Accepted: 06/21/2023] [Indexed: 08/06/2023] Open
Abstract
During cortical development, human basal radial glial cells (bRGCs) are highly capable of sustained self-renewal and neurogenesis. Selective pressures on this cell type may have contributed to the evolution of the human neocortex, leading to an increase in cortical size. bRGCs have enriched expression for Forkhead Box P1 (FOXP1), a transcription factor implicated in neurodevelopmental disorders (NDDs) such as autism spectrum disorder. However, the cell type-specific roles of FOXP1 in bRGCs during cortical development remain unexplored. Here, we examine the requirement for FOXP1 gene expression regulation underlying the production of bRGCs using human brain organoids. We examine a developmental time point when FOXP1 expression is highest in the cortical progenitors, and the bRGCs, in particular, begin to actively produce neurons. With the loss of FOXP1, we show a reduction in the number of bRGCs, as well as reduced proliferation and differentiation of the remaining bRGCs, all of which lead to reduced numbers of excitatory cortical neurons over time. Using single-nuclei RNA sequencing and cell trajectory analysis, we uncover a role for FOXP1 in directing cortical progenitor proliferation and differentiation by regulating key signaling pathways related to neurogenesis and NDDs. Together, these results demonstrate that FOXP1 regulates human-specific features in early cortical development.
Collapse
Affiliation(s)
- Seon Hye E. Park
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
18
|
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
|
19
|
Zangouei AS, Tolue Ghasaban F, Dalili A, Akhlaghipour I, Moghbeli M. MicroRNAs as the pivotal regulators of Forkhead box protein family during gastrointestinal tumor progression and metastasis. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Yan M, Li J, Yan L, Li X, Chen JG. Transcription factor Foxp1 is essential for the induction of choroidal neovascularization. EYE AND VISION 2022; 9:10. [PMID: 35248156 PMCID: PMC8898411 DOI: 10.1186/s40662-022-00281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/13/2022] [Indexed: 12/04/2022]
Abstract
Background The exudative form of age-related macular degeneration (AMD) is characterized by abnormal blood vessel growth, which is stimulated by vascular endothelial growth factor (VEGF) released from retinal pigment epithelium (RPE). The angiogenic behaviors of vascular endothelial cells in vitro depend on forkhead box protein P1 (Foxp1), a transcription repressor widely expressed in human and murine tissues during development. In this study, we aimed to determine whether loss of Foxp1 affects laser-induced choroidal neovascularization (CNV) in mouse. Methods Eye-selective deletion of Foxp1 was obtained by crossing Foxp1flox/flox with Six3-Cre mice. Laser photocoagulation was delivered to six- to eight-week-old mice to induce CNV. The expression of Foxp1 and Cre was determined by immunofluorescence in cryostat sections of the eyes. Fundus fluorescein angiography (FFA), optical coherence tomography (OCT), and B4 isolectin staining were applied to analyze the leakage, bulge height, and area of CNV lesions, respectively. RPE-choroid tissues were isolated for the determination of VEGF and pigment epithelium derived factor (PEDF) by Western blotting. Results Foxp1 was expressed in retinal ganglion cells, RPE, and the choroidal endothelial cells. Laser photocoagulation increased the number of Foxp1+-endothelial cells and induced CNV. Six3-Cre reduced Foxp1 expression in RPE but not the endothelium, leading to a lower level of VEGF in the RPE-choroid. Foxp1 knockout inhibited pathological angiogenesis and vascular leakage of the laser-induced CNV lesions. Conclusions Foxp1 regulates the expression of VEGF in the RPE, and inhibition of Foxp1 could potentially be a novel strategy for the prevention and therapy of neovascularization related to AMD.
Collapse
|
21
|
Transcription factor Foxp1 stimulates angiogenesis in adult rats after myocardial infarction. Cell Death Dis 2022; 8:381. [PMID: 36088337 PMCID: PMC9464245 DOI: 10.1038/s41420-022-01180-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022]
Abstract
Forkhead box protein P1 (FoxP1) is essential for cardiac development and the regulation of neovascularization, but its potential for cardiac angiogenesis has not been explored. This study aims to investigate the angiogenic role of FoxP1 in a rat model of myocardial infarction (MI). Adult male rats were subjected to MI, and Foxp1 was knocked down with lentivirus FoxP1 siRNA. Endothelial cell proliferation, angiogenesis, and cardiac function were also assessed. Cell scratch assay and tubule formation analysis were used to detect the migration ability and tube formation ability of human umbilical vein endothelial cells (HUVECs). Compared with that in the sham group, results showed that the expression of FoxP1 was significantly increased in the MI group. Foxp1 knockdown decreases FoxP1 expression, reduces angiogenesis, and increases collagen deposition. When Foxp1 was knocked down in HUVECs using FoxP1 siRNA lentivirus, cell proliferation, migration, and tube formation abilities decreased significantly. Our study showed that FoxP1 elicits pleiotropic beneficial actions on angiogenesis in the post-MI heart by promoting the proliferation of endothelial cells. FoxP1 should be considered a candidate for therapeutic cardiac angiogenesis.
Collapse
|
22
|
Pirruccello JP, Di Achille P, Nauffal V, Nekoui M, Friedman SF, Klarqvist MDR, Chaffin MD, Weng LC, Cunningham JW, Khurshid S, Roselli C, Lin H, Koyama S, Ito K, Kamatani Y, Komuro I, Jurgens SJ, Benjamin EJ, Batra P, Natarajan P, Ng K, Hoffmann U, Lubitz SA, Ho JE, Lindsay ME, Philippakis AA, Ellinor PT. Genetic analysis of right heart structure and function in 40,000 people. Nat Genet 2022; 54:792-803. [PMID: 35697867 PMCID: PMC10313645 DOI: 10.1038/s41588-022-01090-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/26/2022] [Indexed: 01/29/2023]
Abstract
Congenital heart diseases often involve maldevelopment of the evolutionarily recent right heart chamber. To gain insight into right heart structure and function, we fine-tuned deep learning models to recognize the right atrium, right ventricle and pulmonary artery, measuring right heart structures in 40,000 individuals from the UK Biobank with magnetic resonance imaging. Genome-wide association studies identified 130 distinct loci associated with at least one right heart measurement, of which 72 were not associated with left heart structures. Loci were found near genes previously linked with congenital heart disease, including NKX2-5, TBX5/TBX3, WNT9B and GATA4. A genome-wide polygenic predictor of right ventricular ejection fraction was associated with incident dilated cardiomyopathy (hazard ratio, 1.33 per standard deviation; P = 7.1 × 10-13) and remained significant after accounting for a left ventricular polygenic score. Harnessing deep learning to perform large-scale cardiac phenotyping, our results yield insights into the genetic determinants of right heart structure and function.
Collapse
Affiliation(s)
- James P Pirruccello
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paolo Di Achille
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Victor Nauffal
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mahan Nekoui
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Samuel F Friedman
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marcus D R Klarqvist
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan W Cunningham
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Shaan Khurshid
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Carolina Roselli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Honghuang Lin
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, MA, USA
- Division of Clinical Informatics, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Satoshi Koyama
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Emelia J Benjamin
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, MA, USA
- Department of Medicine, Cardiology and Preventive Medicine Sections, Boston University School of Medicine, Boston, MA, USA
- Epidemiology Department, Boston University School of Public Health, Boston, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pradeep Natarajan
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Udo Hoffmann
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Steven A Lubitz
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer E Ho
- Harvard Medical School, Boston, MA, USA
- CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mark E Lindsay
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Thoracic Aortic Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Patrick T Ellinor
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
23
|
Mitochondrial dysfunction and oxidative stress contribute to cognitive and motor impairment in FOXP1 syndrome. Proc Natl Acad Sci U S A 2022; 119:2112852119. [PMID: 35165191 PMCID: PMC8872729 DOI: 10.1073/pnas.2112852119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 12/22/2022] Open
Abstract
FOXP1 haploinsufficiency underlies cognitive and motor impairments in individuals with FOXP1 syndrome. Here, we show that mice lacking one Foxp1 copy exhibit similar behavioral deficits, which may be caused by striatal dysfunction. Indeed, Foxp1+/− striatal medium spiny neurons display reduced neurite branching, and we show altered mitochondrial biogenesis and dynamics; increased mitophagy; reduced mitochondrial membrane potential, structure, and motility; and elevated oxygen species in the striatum of these animals. As FOXP1 is highly conserved, our data strongly suggest that mitochondrial dysfunction and excessive oxidative stress contribute to the motor and cognitive impairments seen in individuals with FOXP1 syndrome. Thus, mitochondrial homeostasis is critical for normal development and can explain deficits in neurodevelopmental disorders. FOXP1 syndrome caused by haploinsufficiency of the forkhead box protein P1 (FOXP1) gene is a neurodevelopmental disorder that manifests motor dysfunction, intellectual disability, autism, and language impairment. In this study, we used a Foxp1+/− mouse model to address whether cognitive and motor deficits in FOXP1 syndrome are associated with mitochondrial dysfunction and oxidative stress. Here, we show that genes with a role in mitochondrial biogenesis and dynamics (e.g., Foxo1, Pgc-1α, Tfam, Opa1, and Drp1) were dysregulated in the striatum of Foxp1+/− mice at different postnatal stages. Furthermore, these animals exhibit a reduced mitochondrial membrane potential and complex I activity, as well as decreased expression of the antioxidants superoxide dismutase 2 (Sod2) and glutathione (GSH), resulting in increased oxidative stress and lipid peroxidation. These features can explain the reduced neurite branching, learning and memory, endurance, and motor coordination that we observed in these animals. Taken together, we provide strong evidence of mitochondrial dysfunction in Foxp1+/− mice, suggesting that insufficient energy supply and excessive oxidative stress underlie the cognitive and motor impairment in FOXP1 deficiency.
Collapse
|
24
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Forkhead box protein 1 transcriptionally activates sestrin1 to alleviate oxidized low-density lipoprotein-induced inflammation and lipid accumulation in macrophages. Bioengineered 2022; 13:2917-2926. [PMID: 35043753 PMCID: PMC8974195 DOI: 10.1080/21655979.2021.2000228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transcription factor forkhead box protein 1 (FOXP1) has been shown cardiovascular protection. We aimed to analyze the role of FOXP1 in oxidized low-density lipoprotein (ox-LDL)-induced macrophages and its possible regulatory effect on sestrin1 (SESN1) expression. After stimulation with ox-LDL, FOXP1 expression in RAW264.7 cells was evaluated with RT-qPCR and Western blotting. Then, FOXP1 was overexpressed, followed by detection of inflammatory mediator levels using ELISA kits and RT-qPCR. Lipid accumulation was detected with oil red O staining. Additionally, the JASPAR database was used to predict the potential genes that could be transcriptionally regulated by FOXP1. ChIP and luciferase reporter assays were used to verify this combination. To further clarify the regulatory effects of FOXP1 on SESN1 in damage of macrophages triggered by ox-LDL, SESN1 was silenced to determine the inflammation and lipid accumulation under the condition of FOXP1 overexpression. Results indicated that ox-LDL stimulation led to a significant decrease in FOXP1 expression. FOXP1 overexpression notably reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, accompanied by a decreased in phosphorylated NF-κB p65 expression. Besides, FOXP1-upregulation inhibited lipid accumulation and reduced CD36 expression level in RAW264.7 cells upon ox-LDL stimulation. Moreover, results of ChIP and luciferase reporter assays suggested that FOXP1 could transcriptionally regulate SESN1 expression. Further experiments supported that SESN1 silencing restored the inhibitory effects of FOXP1 overexpression on the inflammation and lipid accumulation in RAW264.7 cells exposed to ox-LDL. Collectively, FOXP1 transcriptionally activates SESN1 for the alleviation of ox-LDL-induced inflammation and lipid accumulation in macrophages.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| |
Collapse
|
25
|
Liu X, Yang Y, Song J, Li D, Liu X, Li C, Ma Z, Zhong J, Wang L. Knockdown of forkhead box protein P1 alleviates hypoxia reoxygenation injury in H9c2 cells through regulating Pik3ip1/Akt/eNOS and ROS/mPTP pathway. Bioengineered 2022; 13:1320-1334. [PMID: 35000528 PMCID: PMC8805992 DOI: 10.1080/21655979.2021.2016046] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Forkhead box protein P1 (Foxp1) exerts an extensive array of physiological and pathophysiological impacts on the cardiovascular system. However, the exact function of myocardial Foxp1 in myocardial ischemic reperfusion injury (MIRI) stays largely vague. The hypoxia reoxygenation model of H9c2 cells (the rat ventricular myoblasts) closely mimics myocardial ischemia-reperfusion injury. This report intends to research the effects and mechanisms underlying Foxp1 on H9c2 cells in response to hypoxia (12 h)/reoxygenation (4 h) (HR) stimulation. Expressions of Foxp1 and Phosphatidylinositol 3-kinase interacting protein 1 (Pik3ip1) were both upregulated in ischemia/reperfusion (IR)/HR-induced injury. Stimulation through HR led to marked increases in cellular apoptosis, mitochondrial dysfunction, and superoxide generation in H9c2 cells, which were rescued with knockdown of Foxp1 by siRNA. Silence of Foxp1 depressed expression of Pik3ip1 directly activated the PI3K/Akt/eNOS pathway and promoted nitric oxide (NO) release. Moreover, the knockdown of Foxp1 blunted HR-induced enhancement of reactive oxygen species (ROS) generation, thus alleviating excessive persistence of mitochondrial permeability transition pore (mPTP) opening and decreased mitochondrial apoptosis-associated protein expressions in H9c2 cells. Meanwhile, these cardioprotective effects can be abolished by LY294002, NG-nitro-L-arginine methyl ester (L-NAME), and Atractyloside (ATR), respectively. In summary, our findings indicated that knockdown of Foxp1 prevented HR-induced encouragement of apoptosis and oxidative stress via PI3K/Akt/eNOS signaling activation by targeting Pik3ip1 and improved mitochondrial function by inhibiting ROS-mediated mPTP opening. Inhibition of Foxp1 may be a promising therapeutic avenue for MIRI.
Collapse
Affiliation(s)
- Xinming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yixing Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiawei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Dongjie Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chuang Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zheng Ma
- Department of Cardiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Respiratory Medicine, Beijing, China
| | - Lefeng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Wang J, Rappold GA, Fröhlich H. Disrupted Mitochondrial Network Drives Deficits of Learning and Memory in a Mouse Model of FOXP1 Haploinsufficiency. Genes (Basel) 2022; 13:127. [PMID: 35052467 PMCID: PMC8775322 DOI: 10.3390/genes13010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/16/2022] Open
Abstract
Reduced cognitive flexibility, characterized by restricted interests and repetitive behavior, is associated with atypical memory performance in autism spectrum disorder (ASD), suggesting hippocampal dysfunction. FOXP1 syndrome is a neurodevelopmental disorder characterized by ASD, language deficits, global developmental delay, and mild to moderate intellectual disability. Strongly reduced Foxp1 expression has been detected in the hippocampus of Foxp1+/- mice, a brain region required for learning and memory. To investigate learning and memory performance in these animals, fear conditioning tests were carried out, which showed impaired associative learning compared with wild type (WT) animals. To shed light on the underlying mechanism, we analyzed various components of the mitochondrial network in the hippocampus. Several proteins regulating mitochondrial biogenesis (e.g., Foxo1, Pgc-1α, Tfam) and dynamics (Mfn1, Opa1, Drp1 and Fis1) were significantly dysregulated, which may explain the increased mitophagy observed in the Foxp1+/- hippocampus. The reduced activity of complex I and decreased expression of Sod2 most likely increase the production of reactive oxygen species and the expression of the pre-apoptotic proteins Bcl-2 and Bax in this tissue. In conclusion, we provide evidence that a disrupted mitochondrial network and the resulting oxidative stress in the hippocampus contribute to the altered learning and cognitive impairment in Foxp1+/- mice, suggesting that similar alterations also play a major role in patients with FOXP1 syndrome.
Collapse
Affiliation(s)
- Jing Wang
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
| | - Gudrun A. Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
- Interdisciplinary Center for Neurosciences, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Henning Fröhlich
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
| |
Collapse
|
27
|
Hu J, Liu X, Tang Y. HMGB1/Foxp1 regulates hypoxia-induced inflammatory response in macrophages. Cell Biol Int 2021; 46:265-277. [PMID: 34816539 DOI: 10.1002/cbin.11728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/14/2021] [Accepted: 11/13/2021] [Indexed: 01/04/2023]
Abstract
Forkhead box protein P1 (Foxp1) is a kind of tumor suppressor gene, and the role of Foxp1 in the macrophages of myocardial infarction (MI) has not been studied yet. Here, we verified the role of the transcription factor high mobility group box 1 (HMGB1) and its target gene Foxp1 in the inflammatory response. In this study, the key genes HMGB1 and Foxp1 in the macrophages of mouse MI model were screened out through single-cell transcriptome analysis of GSE136088 (GEO database). In vitro experiment indicated that hypoxia induced the inflammatory response in RAW264.7 macrophages, promoted the secretion of inflammatory factors (tumor necrosis factor α [TNF-α], interleukin 6 [IL-6], and IL-1β) and the activation of NLRP3 inflammasome (NLRP3, ASC, and pro-caspase-1). Meanwhile, HMGB1 increased while Foxp1 decreased in hypoxia-treated RAW264.7 macrophages. HMGB1 bound to the upstream promoter region of Foxp1 as demonstrated by the dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP)-quantitative polymerase chain reaction (qPCR) and agarose gel electrophoresis. As a transcription factor, HMGB1 regulated Foxp1 expression. The secretion of inflammatory factors and the expression of NLRP3 inflammasome protein were changed when the expression of HMGB1 and Foxp1 was regulated in the hypoxia-treated RAW264.7 macrophages. This study verified that HMGB1 could aggravate the hypoxia-treated inflammatory response of macrophages through downregulating Foxp1, which not only provides evidence to support the role of HMGB1/Foxp1 in macrophages but also offers another angle for the treatment of MI.
Collapse
Affiliation(s)
- Jing Hu
- Department of Cardiovascular, The First Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Xiaojun Liu
- Department of Cardiovascular, The First Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Yu Tang
- Department of Cardiovascular, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
28
|
Detection of Novel Potential Regulators of Stem Cell Differentiation and Cardiogenesis through Combined Genome-Wide Profiling of Protein-Coding Transcripts and microRNAs. Cells 2021; 10:cells10092477. [PMID: 34572125 PMCID: PMC8469649 DOI: 10.3390/cells10092477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
In vitro differentiation of embryonic stem cells (ESCs) provides a convenient basis for the study of microRNA-based gene regulation that is relevant for early cardiogenic processes. However, to which degree insights gained from in vitro differentiation models can be readily transferred to the in vivo system remains unclear. In this study, we profiled simultaneous genome-wide measurements of mRNAs and microRNAs (miRNAs) of differentiating murine ESCs (mESCs) and integrated putative miRNA-gene interactions to assess miRNA-driven gene regulation. To identify interactions conserved between in vivo and in vitro, we combined our analysis with a recent transcriptomic study of early murine heart development in vivo. We detected over 200 putative miRNA-mRNA interactions with conserved expression patterns that were indicative of gene regulation across the in vitro and in vivo studies. A substantial proportion of candidate interactions have been already linked to cardiogenesis, supporting the validity of our approach. Notably, we also detected miRNAs with expression patterns that closely resembled those of key developmental transcription factors. The approach taken in this study enabled the identification of miRNA interactions in in vitro models with potential relevance for early cardiogenic development. Such comparative approaches will be important for the faithful application of stem cells in cardiovascular research.
Collapse
|
29
|
Lin SZ, Zhou XY, Wang WQ, Jiang K. Autism with dysphasia accompanied by mental retardation caused by FOXP1 exon deletion: A case report. World J Clin Cases 2021; 9:6858-6866. [PMID: 34447835 PMCID: PMC8362507 DOI: 10.12998/wjcc.v9.i23.6858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/24/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Forkhead box protein 1 (FOXP1) (OMIM: 605515) at chromosomal region 3p14.1 plays an important regulatory role in cell development and functions by regulating genetic expression. Earlier studies have suggested that FOXP1, an oncogene, is capable of initiating tumorigenicity depending on the cell type. FOXP1 also plays an important role in regulating the cell development and functions of the immune system, e.g., regulating B-cell maturation and mononuclear phagocyte differentiation, and in the occurrence and development of various immune diseases. The mRNA of this gene is widely expressed in humans, and its differential expression is related to numerous diseases.
CASE SUMMARY A 5-year-old boy mainly presented with attention deficit and hyperactivity disorder and developmental retardation accompanied by gait instability and abnormal facial features (low-set ears). DNA samples were extracted from the child’s and his parents’ peripheral blood to detect whole-exome sequences and whole-genome copy number variations. Results revealed heterozygous deletions of exon 6-21 of FOXP1 gene in the child. Physical examination upon admission showed that the child was generally in good condition, had a moderate nutritional status, a slightly slow response to external stimuli, equally large and equally round bilateral pupils, was sensitive to light reflection, and had poor eye contact and joint attention. He had no meaningful utterance and could not pronounce words properly. He was able to use gestures to simply express his thoughts, to perform simple actions, and to listen to instructions. He had no rash, cafe-au-lait macules, or depigmentation spots. He had thick black hair and low-set ears. He had highly sensitive skin, especially on his face and palms. He had no abnormal palm fingerprint. Cardiopulmonary and abdominal examinations revealed no abnormalities. He had normal limb muscle strength and tension. He showed normal tendon reflexes of both knees. His bilateral Babinski and meningeal irritation signs were negative. He had a normal male vulva.
CONCLUSION We report the characteristic features of autism with dysphasia accompanied by mental retardation caused by FOXP1 exon deletion. This study provides a molecular basis for etiological diagnosis and treatment of the child, as well as for genetic counseling for the pedigree.
Collapse
Affiliation(s)
- Shuang-Zhu Lin
- Department of Diagnosis and Treatment Center for Children, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Xin-Yu Zhou
- Department of Diagnosis and Treatment Center for Children, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Wan-Qi Wang
- Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Kai Jiang
- Department of Diagnosis and Treatment Center for Children, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| |
Collapse
|
30
|
Forkhead Transcription Factors in Health and Disease. Trends Genet 2021; 37:460-475. [DOI: 10.1016/j.tig.2020.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
|
31
|
Lozano R, Gbekie C, Siper PM, Srivastava S, Saland JM, Sethuram S, Tang L, Drapeau E, Frank Y, Buxbaum JD, Kolevzon A. FOXP1 syndrome: a review of the literature and practice parameters for medical assessment and monitoring. J Neurodev Disord 2021; 13:18. [PMID: 33892622 PMCID: PMC8066957 DOI: 10.1186/s11689-021-09358-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/25/2021] [Indexed: 11/14/2022] Open
Abstract
FOXP1 syndrome is a neurodevelopmental disorder caused by mutations or deletions that disrupt the forkhead box protein 1 (FOXP1) gene, which encodes a transcription factor important for the early development of many organ systems, including the brain. Numerous clinical studies have elucidated the role of FOXP1 in neurodevelopment and have characterized a phenotype. FOXP1 syndrome is associated with intellectual disability, language deficits, autism spectrum disorder, hypotonia, and congenital anomalies, including mild dysmorphic features, and brain, cardiac, and urogenital abnormalities. Here, we present a review of human studies summarizing the clinical features of individuals with FOXP1 syndrome and enlist a multidisciplinary group of clinicians (pediatrics, genetics, psychiatry, neurology, cardiology, endocrinology, nephrology, and psychology) to provide recommendations for the assessment of FOXP1 syndrome.
Collapse
Affiliation(s)
- Reymundo Lozano
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Catherine Gbekie
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paige M Siper
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shubhika Srivastava
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeffrey M Saland
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Children's Heart Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swathi Sethuram
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lara Tang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elodie Drapeau
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yitzchak Frank
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Neyroud D, Nosacka RL, Callaway CS, Trevino JG, Hu H, Judge SM, Judge AR. FoxP1 is a transcriptional repressor associated with cancer cachexia that induces skeletal muscle wasting and weakness. J Cachexia Sarcopenia Muscle 2021; 12:421-442. [PMID: 33527776 PMCID: PMC8061399 DOI: 10.1002/jcsm.12666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/05/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Skeletal muscle wasting is a devastating consequence of cancer that affects up to 80% of cancer patients and associates with reduced survival. Herein, we investigated the biological significance of Forkhead box P1 (FoxP1), a transcriptional repressor that we demonstrate is up-regulated in skeletal muscle in multiple models of cancer cachexia and in cachectic cancer patients. METHODS Inducible, skeletal muscle-specific FoxP1 over-expressing (FoxP1iSkmTg/Tg ) mice were generated through crossing conditional Foxp1a transgenic mice with HSA-MCM mice that express tamoxifen-inducible Cre recombinase under control of the skeletal muscle actin promoter. To determine the requirement of FoxP1 for cancer-induced skeletal muscle wasting, FoxP1-shRNA was packaged and targeted to muscles using AAV9 delivery prior to inoculation of mice with Colon-26 Adenocarcinoma (C26) cells. RESULTS Up-regulation of FoxP1 in adult skeletal muscle was sufficient to induce features of cachexia, including 15% reduction in body mass (P < 0.05), and a 16-27% reduction in skeletal muscle mass (P < 0.05) that was characterized by a 20% reduction in muscle fibre cross-sectional area of type IIX/B muscle fibres (P = 0.020). Skeletal muscles from FoxP1iSkmTg/Tg mice also showed significant damage and myopathy characterized by the presence of centrally nucleated myofibres, extracellular matrix expansion, and were 19-26% weaker than controls (P < 0.05). Transcriptomic analysis revealed FoxP1 as a potent transcriptional repressor of skeletal muscle gene expression, with enrichment of pathways related to skeletal muscle structure and function, growth signalling, and cell quality control. Because FoxP1 functions, at least in part, as a transcriptional repressor through its interaction with histone deacetylase proteins, we treated FoxP1iSkmTg/Tg mice with Trichostatin A, and found that this completely prevented the loss of muscle mass (p = 0.007) and fibre atrophy (P < 0.001) in the tibialis anterior. In the context of cancer, FoxP1 knockdown blocked the cancer-induced repression of myocyte enhancer factor 2 (MEF2)-target genes critical to muscle differentiation and repair, improved muscle ultrastructure, and attenuated muscle fibre atrophy by 50% (P < 0.05). CONCLUSIONS In summary, we identify FoxP1 as a novel repressor of skeletal muscle gene expression that is increased in cancer cachexia, whose up-regulation is sufficient to induce skeletal muscle wasting and weakness, and required for the normal wasting response to cancer.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | | | | | | | - Hui Hu
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Sarah M. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | - Andrew R. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
33
|
Pearson CA, Moore DM, Tucker HO, Dekker JD, Hu H, Miquelajáuregui A, Novitch BG. Foxp1 Regulates Neural Stem Cell Self-Renewal and Bias Toward Deep Layer Cortical Fates. Cell Rep 2021; 30:1964-1981.e3. [PMID: 32049024 DOI: 10.1016/j.celrep.2020.01.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
The laminar architecture of the mammalian neocortex depends on the orderly generation of distinct neuronal subtypes by apical radial glia (aRG) during embryogenesis. Here, we identify critical roles for the autism risk gene Foxp1 in maintaining aRG identity and gating the temporal competency for deep-layer neurogenesis. Early in development, aRG express high levels of Foxp1 mRNA and protein, which promote self-renewing cell divisions and deep-layer neuron production. Foxp1 levels subsequently decline during the transition to superficial-layer neurogenesis. Sustained Foxp1 expression impedes this transition, preserving a population of cells with aRG identity throughout development and extending the early neurogenic period into postnatal life. FOXP1 expression is further associated with the initial formation and expansion of basal RG (bRG) during human corticogenesis and can promote the formation of cells exhibiting characteristics of bRG when misexpressed in the mouse cortex. Together, these findings reveal broad functions for Foxp1 in cortical neurogenesis.
Collapse
Affiliation(s)
- Caroline Alayne Pearson
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Destaye M Moore
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Haley O Tucker
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Joseph D Dekker
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Hui Hu
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Amaya Miquelajáuregui
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00911, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
The Drosophila Forkhead/Fox transcription factor Jumeau mediates specific cardiac progenitor cell divisions by regulating expression of the kinesin Nebbish. Sci Rep 2021; 11:3221. [PMID: 33547352 PMCID: PMC7864957 DOI: 10.1038/s41598-021-81894-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022] Open
Abstract
Forkhead (Fkh/Fox) domain transcription factors (TFs) mediate multiple cardiogenic processes in both mammals and Drosophila. We showed previously that the Drosophila Fox gene jumeau (jumu) controls three categories of cardiac progenitor cell division—asymmetric, symmetric, and cell division at an earlier stage—by regulating Polo kinase activity, and mediates the latter two categories in concert with the TF Myb. Those observations raised the question of whether other jumu-regulated genes also mediate all three categories of cardiac progenitor cell division or a subset thereof. By comparing microarray-based expression profiles of wild-type and jumu loss-of-function mesodermal cells, we identified nebbish (neb), a kinesin-encoding gene activated by jumu. Phenotypic analysis shows that neb is required for only two categories of jumu-regulated cardiac progenitor cell division: symmetric and cell division at an earlier stage. Synergistic genetic interactions between neb, jumu, Myb, and polo and the rescue of jumu mutations by ectopic cardiac mesoderm-specific expression of neb demonstrate that neb is an integral component of a jumu-regulated subnetwork mediating cardiac progenitor cell divisions. Our results emphasize the central role of Fox TFs in cardiogenesis and illustrate how a single TF can utilize different combinations of other regulators and downstream effectors to control distinct developmental processes.
Collapse
|
35
|
Li Y, Li X, Wang L, Han N, Yin G. miR-375-3p contributes to hypoxia-induced apoptosis by targeting forkhead box P1 (FOXP1) and Bcl2 like protein 2 (Bcl2l2) in rat cardiomyocyte h9c2 cells. Biotechnol Lett 2020; 43:353-367. [PMID: 33128129 DOI: 10.1007/s10529-020-03013-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
miRNAs have been pointed to play critical role in the development of congenital heart disease (CHD). miRNA-375-3p (miR-375-3p) was involved in cardiac dysfunction and cardiogenesis. However, no prior study had established a therapeutic role of miR-375-3p in CHD. We intended to investigate the effect and mechanism of miR-375-3p on apoptosis in hypoxic cardiomyocytes in vitro. Expression of miR-375-3p, forkhead box P1 (FOXP1) and Bcl2 like protein 2 (Bcl2l2) was detected using real-time quantitative PCR and western blot. Apoptosis was measured with MTT assay, flow cytometry and caspase-3 activity assay. The potential target binding between miR-375-3p and FOXP1/Bcl2l2 was predicted on DianaTools, and was validated by luciferase reporter assay and RNA pull-down assay. As a result, miR-375-3p was upregulated and FOXP1/Bcl2l2 was downregulated in maternal serum of women with fetal CHD and hypoxia-induced rat cardiomyocyte h9c2 cells. Hypoxia induced apoptosis rate elevation, caspase-3 activity promotion and viability inhibition in h9c2 cells; overexpression of miR-375-3p promoted, whereas knockdown of miR-375-3p antagonized hypoxia-induced effects in h9c2 cells. In addition, miR-375-3p was validated to negatively regulate FOXP1 and Bcl2l2 expression through target binding, and silencing of FOXP1 and Bcl2l2 could independently abate the anti-apoptosis role of miR-375-3p knockdown in hypoxic h9c2 cells. Collectively, blocking miR-375-3p suppressed hypoxia-evoked apoptosis of cardiomyocytes by targeting and upregulating FOXP1 and Bcl2l2. Our results might suggest maternal serum miR-375-3p as a potential biomarker for prenatal detection of fetal CHD.
Collapse
Affiliation(s)
- Yuefan Li
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China
| | - Xiaofei Li
- Department of Acupuncture, Qingdao Central Hospital, Qingdao, 266042, Shandong, China
| | - Ling Wang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Na Han
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China
| | - Gang Yin
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China.
| |
Collapse
|
36
|
Li X, Han X, Tu X, Zhu D, Feng Y, Jiang T, Yang Y, Qu J, Chen JG. An Autism-Related, Nonsense Foxp1 Mutant Induces Autophagy and Delays Radial Migration of the Cortical Neurons. Cereb Cortex 2020; 29:3193-3208. [PMID: 30124790 DOI: 10.1093/cercor/bhy185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/16/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that has a strong genetic component. Disruptions of FOXP1, a transcription factor expressed in the developing cerebral cortex, were associated with ASD. FOXP1(R525X) is a de novo heterozygous mutation found in patients with autism and severe mental retardation. To explore the neuronal basis of FOXP1(R525X) in ASD, we created Foxp1(R521X), a mouse homolog of the human variant. Ectopic expression of Foxp1(R521X) led to cytoplasmic aggregates and activated macroautophagy in neuroblastoma N2a cells and the developing neuronal cells. Cortical neurons expressing Foxp1(R521X) exhibited delayed migration and altered dendritic morphology. As a control, mutant Y435X that was expressed diffusively in the cytoplasm did not induce autophagy and migration delay in the cortex. The embryonic cortical cells had a minimal activity of nonsense-mediated mRNA decay (NMD) as assayed by a splicing-dependent NMD reporter. We hypothesize that the developing neuronal cells use autophagy but not NMD as a safeguard mechanism against nonsense mutant aggregates, resulting in impairment of the cortical development. This study suggests a novel mechanism other than heterozygous loss of FOXP1 for the development of ASD and may advance our understanding of the complex relationships between gene mutation and the related psychiatric disorders.
Collapse
Affiliation(s)
- Xue Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Xin Han
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Xiaomeng Tu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Dan Zhu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Yue Feng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Tian Jiang
- Research Center for Translational Medicine, the Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, PR China
| | - Youping Yang
- Research Center for Translational Medicine, the Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, PR China
| | - Jia Qu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| | - Jie-Guang Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.,State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, PR China.,Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang, PR China
| |
Collapse
|
37
|
Co M, Anderson AG, Konopka G. FOXP transcription factors in vertebrate brain development, function, and disorders. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e375. [PMID: 31999079 PMCID: PMC8286808 DOI: 10.1002/wdev.375] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
FOXP transcription factors are an evolutionarily ancient protein subfamily coordinating the development of several organ systems in the vertebrate body. Association of their genes with neurodevelopmental disorders has sparked particular interest in their expression patterns and functions in the brain. Here, FOXP1, FOXP2, and FOXP4 are expressed in distinct cell type-specific spatiotemporal patterns in multiple regions, including the cortex, hippocampus, amygdala, basal ganglia, thalamus, and cerebellum. These varied sites and timepoints of expression have complicated efforts to link FOXP1 and FOXP2 mutations to their respective developmental disorders, the former affecting global neural functions and the latter specifically affecting speech and language. However, the use of animal models, particularly those with brain region- and cell type-specific manipulations, has greatly advanced our understanding of how FOXP expression patterns could underlie disorder-related phenotypes. While many questions remain regarding FOXP expression and function in the brain, studies to date have illuminated the roles of these transcription factors in vertebrate brain development and have greatly informed our understanding of human development and disorders. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Marissa Co
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Ashley G Anderson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
38
|
Gu C, Luo Y, Zhang S, Xu J, Zhang J, Ju H, Liu J, Zhang L, Zhang Y, Wu L, Xie E, Xu T, Pan S. MAb NJ001 inhibits lung adenocarcinoma invasiveness by directly regulating TIMP-3 promoter activity via FOXP1 binding sites. Thorac Cancer 2020; 11:2630-2638. [PMID: 32744429 PMCID: PMC7471035 DOI: 10.1111/1759-7714.13593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/27/2022] Open
Abstract
Background Previously, we developed a monoclonal antibody (mAb) NJ001 that binds to the antigen SP70 in human non‐small cell lung cancer (NSCLC) cells and showed it could inhibit lung adenocarcinoma (AD) growth. Here, we investigated the effect and mechanisms of NJ001 in lung AD metastasis. Methods Human lung AD cells (SPC‐A1 and A549) were treated with different concentrations of mAb NJ001, and the effects of NJ001 on cell migration and invasive activity were investigated using wound‐healing and Matrigel assays, respectively. The molecular mechanism of this inhibition was explored by microarrays, qRT‐PCR, western blot, luciferase assays and electrophoretic mobility shift assays (EMSA). Results MAb NJ001 markedly suppressed lung AD cell migration; and the invasiveness of SPC‐A1 and A549 cells treated with mAb NJ001 was diminished by 65%. Tissue inhibitor of matrix metalloproteinase‐3 (TIMP‐3) was highly expressed in SPC‐A1 cells treated with mAb NJ001, whereas knockdown of TIMP‐3 by shRNA significantly increased SPC‐A1 and A549 invasiveness. MAb NJ001 affects lung AD by inhibiting TIMP‐3 through direct transcriptional regulation of FOXP1 binding sites in the TIMP‐3 promoter region, as shown in luciferase assays and EMSA. Conclusions MAb NJ001 inhibits invasiveness and metastasis in lung AD through the FOXP1 binding sites in the TIMP‐3 promoter region. It may have clinical applications in preventing and treating metastatic lung AD.
Collapse
Affiliation(s)
- Chunrong Gu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Ying Luo
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Shichang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Jian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Jiexin Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Huanyu Ju
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Jingping Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Lixia Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Yan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Lei Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Erfu Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Ting Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Shiyang Pan
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| |
Collapse
|
39
|
Liu XM, Du SL, Miao R, Wang LF, Zhong JC. Targeting the forkhead box protein P1 pathway as a novel therapeutic approach for cardiovascular diseases. Heart Fail Rev 2020; 27:345-355. [PMID: 32648149 DOI: 10.1007/s10741-020-09992-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide and encompasses diverse diseases of the vasculature, myocardium, cardiac electrical circuit, and cardiac development. Forkhead box protein P1 (Foxp1) is a large multi-domain transcriptional regulator belonging to the Fox family with winged helix DNA-binding protein, which plays critical roles in cardiovascular homeostasis and disorders. The broad distribution of Foxp1 and alternative splicing isoforms implicate its distinct functions in diverse cardiac and vascular cells and tissue types. Foxp1 is essential for diverse biological processes and has been shown to regulate cellular proliferation, apoptosis, oxidative stress, fibrosis, angiogenesis, cardiovascular remodeling, and dysfunction. Notably, both loss-of-function and gain-of-function approaches have defined critical roles of Foxp1 in CVD. Genetic deletion of Foxp1 results in pathological cardiac remodeling, exacerbation of atherosclerotic lesion formation, prolonged occlusive thrombus formation, severe cardiac defects, and embryo death. In contrast, activation of Foxp1 performs a wide range of physiological effects, including cell growth, hypertrophy, differentiation, angiogenesis, and cardiac development. More importantly, Foxp1 exerts anti-inflammatory and anti-atherosclerotic effects in controlling coronary thrombus formation and myocardial infarction (MI). Thus, targeting for Foxp1 signaling has emerged as a pre-warning biomarker and a novel therapeutic approach against progression of CVD, and an increased understanding of cardiovascular actions of the Foxp1 signaling will help to develop effective interventions. In this review, we focus on the diverse actions and underlying mechanisms of Foxp1 highlighting its roles in CVD, including heart failure, MI, atherosclerosis, congenital heart defects, and atrial fibrillation.
Collapse
Affiliation(s)
- Xin-Ming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Sheng-Li Du
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ran Miao
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Le-Feng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China. .,Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
40
|
MiR-195 enhances cardiomyogenic differentiation of the proepicardium/septum transversum by Smurf1 and Foxp1 modulation. Sci Rep 2020; 10:9334. [PMID: 32518241 PMCID: PMC7283354 DOI: 10.1038/s41598-020-66325-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular development is a complex developmental process in which multiple cell lineages are involved, namely the deployment of first and second heart fields. Beside the contribution of these cardiogenic fields, extracardiac inputs to the developing heart are provided by the migrating cardiac neural crest cells and the proepicardial derived cells. The proepicardium (PE) is a transitory cauliflower-like structure located between the cardiac and hepatic primordia. The PE is constituted by an internal mesenchymal component surrounded by an external epithelial lining. With development, cells derived from the proepicardium migrate to the neighboring embryonic heart and progressive cover the most external surface, leading to the formation of the embryonic epicardium. Experimental evidence in chicken have nicely demonstrated that epicardial derived cells can distinctly contribute to fibroblasts, endothelial and smooth muscle cells. Surprisingly, isolation of the developing PE anlage and ex vivo culturing spontaneously lead to differentiation into beating cardiomyocytes, a process that is enhanced by Bmp but halted by Fgf administration. In this study we provide a comprehensive characterization of the developmental expression profile of multiple microRNAs during epicardial development in chicken. Subsequently, we identified that miR-125, miR-146, miR-195 and miR-223 selectively enhance cardiomyogenesis both in the PE/ST explants as well as in the embryonic epicardium, a Smurf1- and Foxp1-driven process. In addition we identified three novel long non-coding RNAs with enhanced expression in the PE/ST, that are complementary regulated by Bmp and Fgf administration and well as by microRNAs that selectively promote cardiomyogenesis, supporting a pivotal role of these long non coding RNAs in microRNA-mediated cardiomyogenesis of the PE/ST cells.
Collapse
|
41
|
Daou R, Beißbarth T, Wingender E, Gültas M, Haubrock M. Constructing temporal regulatory cascades in the context of development and cell differentiation. PLoS One 2020; 15:e0231326. [PMID: 32275727 PMCID: PMC7147753 DOI: 10.1371/journal.pone.0231326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/20/2020] [Indexed: 12/02/2022] Open
Abstract
Cell differentiation is a complex process orchestrated by sets of regulators precisely appearing at certain time points, resulting in regulatory cascades that affect the expression of broader sets of genes, ending up in the formation of different tissues and organ parts. The identification of stage-specific master regulators and the mechanism by which they activate each other is a key to understanding and controlling differentiation, particularly in the fields of tissue regeneration and organoid engineering. Here we present a workflow that combines a comprehensive general regulatory network based on binding site predictions with user-provided temporal gene expression data, to generate a a temporally connected series of stage-specific regulatory networks, which we call a temporal regulatory cascade (TRC). A TRC identifies those regulators that are unique for each time point, resulting in a cascade that shows the emergence of these regulators and regulatory interactions across time. The model was implemented in the form of a user-friendly, visual web-tool, that requires no expert knowledge in programming or statistics, making it directly usable for life scientists. In addition to generating TRCs the tool links multiple interactive visual workflows, in which a user can track and investigate further different regulators, target genes, and interactions, directing the tool along the way into biologically sensible results based on the given dataset. We applied the TRC model on two different expression datasets, one based on experiments conducted on human induced pluripotent stem cells (hiPSCs) undergoing differentiation into mature cardiomyocytes and the other based on the differentiation of H1-derived human neuronal precursor cells. The model was successful in identifying previously known and new potential key regulators, in addition to the particular time points with which these regulators are associated, in cardiac and neural development.
Collapse
Affiliation(s)
- Rayan Daou
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Edgar Wingender
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Science, Georg-August University, Goettingen, Niedersachsen, Germany
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Goettingen, Niedersachsen, Germany
| | - Martin Haubrock
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
- * E-mail:
| |
Collapse
|
42
|
Li W, Pozzo-Miller L. Dysfunction of the corticostriatal pathway in autism spectrum disorders. J Neurosci Res 2019; 98:2130-2147. [PMID: 31758607 DOI: 10.1002/jnr.24560] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
The corticostriatal pathway that carries sensory, motor, and limbic information to the striatum plays a critical role in motor control, action selection, and reward. Dysfunction of this pathway is associated with many neurological and psychiatric disorders. Corticostriatal synapses have unique features in their cortical origins and striatal targets. In this review, we first describe axonal growth and synaptogenesis in the corticostriatal pathway during development, and then summarize the current understanding of the molecular bases of synaptic transmission and plasticity at mature corticostriatal synapses. Genes associated with autism spectrum disorder (ASD) have been implicated in axonal growth abnormalities, imbalance of the synaptic excitation/inhibition ratio, and altered long-term synaptic plasticity in the corticostriatal pathway. Here, we review a number of ASD-associated high-confidence genes, including FMR1, KMT2A, GRIN2B, SCN2A, NLGN1, NLGN3, MET, CNTNAP2, FOXP2, TSHZ3, SHANK3, PTEN, CHD8, MECP2, DYRK1A, RELN, FOXP1, SYNGAP1, and NRXN, and discuss their relevance to proper corticostriatal function.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
43
|
Liu P, Huang S, Ling S, Xu S, Wang F, Zhang W, Zhou R, He L, Xia X, Yao Z, Fan Y, Wang N, Hu C, Zhao X, Tucker HO, Wang J, Guo X. Foxp1 controls brown/beige adipocyte differentiation and thermogenesis through regulating β3-AR desensitization. Nat Commun 2019; 10:5070. [PMID: 31699980 PMCID: PMC6838312 DOI: 10.1038/s41467-019-12988-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/02/2019] [Indexed: 01/08/2023] Open
Abstract
β-Adrenergic receptor (β-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the β3-adrenergic receptor (β3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses β3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity. Beta3-adrenergic receptor (b3-AR) signaling in response to cold activates adipose tissue thermogenesis. Here the authors identify the transcription factor FoxP1 as a direct negative regulator of b3-AR expression and show that loss of FoxP1 leads to enhanced development of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Pei Liu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Sixia Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shifeng Ling
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuqin Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fuhua Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Congxia Hu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaodong Zhao
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xizhi Guo
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
44
|
Gastrointestinal dysfunction in autism displayed by altered motility and achalasia in Foxp1 +/- mice. Proc Natl Acad Sci U S A 2019; 116:22237-22245. [PMID: 31611379 DOI: 10.1073/pnas.1911429116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal dysfunctions in individuals with autism spectrum disorder are poorly understood, although they are common among this group of patients. FOXP1 haploinsufficiency is characterized by autistic behavior, language impairment, and intellectual disability, but feeding difficulties and gastrointestinal problems have also been reported. Whether these are primary impairments, the result of altered eating behavior, or side effects of psychotropic medication remains unclear. To address this question, we investigated Foxp1 +/- mice reflecting FOXP1 haploinsufficiency. These animals show decreased body weight and altered feeding behavior with reduced food and water intake. A pronounced muscular atrophy was detected in the esophagus and colon, caused by reduced muscle cell proliferation. Nitric oxide-induced relaxation of the lower esophageal sphincter was impaired and achalasia was confirmed in vivo by manometry. Foxp1 targets (Nexn, Rbms3, and Wls) identified in the brain were dysregulated in the adult Foxp1 +/- esophagus. Total gastrointestinal transit was significantly prolonged due to impaired colonic contractility. Our results have uncovered a previously unknown dysfunction (achalasia and impaired gut motility) that explains the gastrointestinal disturbances in patients with FOXP1 syndrome, with potential wider relevance for autism.
Collapse
|
45
|
Feng X, Xiong W, Yuan M, Zhan J, Zhu X, Wei Z, Chen X, Cheng X. Down-regulated microRNA-183 mediates the Jak/Stat signaling pathway to attenuate hippocampal neuron injury in epilepsy rats by targeting Foxp1. Cell Cycle 2019; 18:3206-3222. [PMID: 31571517 DOI: 10.1080/15384101.2019.1671717] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recently, the impacts of microRNAs (miRNAs) have been identified in epilepsy (EP), this study was designed to assess the role of miR-183 in hippocampal neuron injury in EP. Rat EP models were established by injected with lithium-pilocarpine. The pathological observation of rats' hippocampus sections was conducted. Expression of miR-183, Foxp1, Jak1, Stat1, and Stat3 in rats' hippocampal tissues was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. The proliferation ability and the apoptosis of the rats' neurons were measured. Furthermore, the target relation between miR-183 and Foxp1 was determined by bioinformatics analysis and dual-luciferase gene reporter assay. The levels of miR-183, Jak1, Stat1, and Stat3 were elevated, and the expression of Foxp1 was declined in EP rats' hippocampal tissues. Inhibited miR-183 could up-regulate Foxp1, inhibited miR-183 together with up-regulated Foxp1 could repress hippocampal neuron injury, promote neuron proliferation, suppress neuron apoptosis, and inactivate the Jak/Stat signaling pathway, resulting in an attenuation of EP progression. Moreover, down-regulated Foxp1 could reverse the attenuation of EP progression which was contributed by inhibited miR-183. Our study implies that inhibited miR-183 could up-regulate Foxp1, resulting in an inactivation of the Jak/Stat signaling pathway and promotion of neuron proliferation, as well as inhibition of apoptosis of hippocampal neurons in EP rats, by which the hippocampal neuron injury and EP progression could be repressed.
Collapse
Affiliation(s)
- Xiangyong Feng
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Wei Xiong
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Mingqiong Yuan
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Jian Zhan
- Department of Neurology, The Second Affiliated Hospital of Zunyi Medical University , Zunyi , Guizhou , PR. China
| | - Xiankun Zhu
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Zhijie Wei
- Department of Neurology, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Xidong Chen
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| | - Xianbing Cheng
- Department of Rehabilitation Medicine, Affiliated hospital of zunyi medical university , Zunyi , Guizhou , PR. China
| |
Collapse
|
46
|
Mulvaney EP, O'Sullivan ÁG, Eivers SB, Reid HM, Kinsella BT. Differential expression of the TPα and TPβ isoforms of the human T Prostanoid receptor during chronic inflammation of the prostate: Role for FOXP1 in the transcriptional regulation of TPβ during monocyte-macrophage differentiation. Exp Mol Pathol 2019; 110:104277. [PMID: 31271729 DOI: 10.1016/j.yexmp.2019.104277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/21/2019] [Accepted: 06/22/2019] [Indexed: 11/16/2022]
Abstract
Inflammation is linked to prostate cancer (PCa) and to other diseases of the prostate. The prostanoid thromboxane (TX)A2 is a pro-inflammatory mediator implicated in several prostatic diseases, including PCa. TXA2 signals through the TPα and TPβ isoforms of the T Prostanoid receptor (TP) which exhibit several functional differences and transcriptionally regulated by distinct promoters Prm1 and Prm3, respectively, within the TBXA2R gene. This study examined the expression of TPα and TPβ in inflammatory infiltrates within human prostate tissue. Strikingly, TPβ expression was detected in 94% of infiltrates, including in B- and T-lymphocytes and macrophages. In contrast, TPα was more variably expressed and, where present, expression was mainly confined to macrophages. To gain molecular insight into these findings, expression of TPα and TPβ was evaluated as a function of monocyte-to-macrophage differentiation in THP-1 cells. Expression of both TPα and TPβ was upregulated following phorbol-12-myristate-13-acetate (PMA)-induced differentiation of monocytic THP-1 to their macrophage lineage. Furthermore, FOXP1, an essential transcriptional regulator down-regulated during monocyte-to-macrophage differentiation, was identified as a key trans-acting factor regulating TPβ expression through Prm3 in THP-1 cells. Knockdown of FOXP1 increased TPβ, but not TPα, expression in THP-1 cells, while genetic reporter and chromatin immunoprecipitation (ChIP) analyses established that FOXP1 exerts its repressive effect on TPβ through binding to four cis-elements within Prm3. Collectively, FOXP1 functions as a transcriptional repressor of TPβ in monocytes. This repression is lifted in differentiated macrophages, allowing for upregulation of TPβ expression and possibly accounting for the prominent expression of TPβ in prostate tissue-resident macrophages.
Collapse
Affiliation(s)
- Eamon P Mulvaney
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Áine G O'Sullivan
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sarah B Eivers
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Helen M Reid
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - B Therese Kinsella
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; ATXA Therapeutics Limited, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
47
|
Liu J, Zhuang T, Pi J, Chen X, Zhang Q, Li Y, Wang H, Shen Y, Tomlinson B, Chan P, Yu Z, Cheng Y, Zheng X, Reilly M, Morrisey E, Zhang L, Liu Z, Zhang Y. Endothelial Forkhead Box Transcription Factor P1 Regulates Pathological Cardiac Remodeling Through Transforming Growth Factor-β1-Endothelin-1 Signal Pathway. Circulation 2019; 140:665-680. [PMID: 31177814 DOI: 10.1161/circulationaha.119.039767] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathological cardiac fibrosis and hypertrophy, the common features of left ventricular remodeling, often progress to heart failure. Forkhead box transcription factor P1 (Foxp1) in endothelial cells (ECs) has been shown to play an important role in heart development. However, the effect of EC-Foxp1 on pathological cardiac remodeling has not been well clarified. This study aims to determine the role of EC-Foxp1 in pathological cardiac remodeling and the underlying mechanisms. METHODS Foxp1 EC-specific loss-of-function and gain-of-function mice were generated, and an angiotensin II infusion or a transverse aortic constriction operation mouse model was used to study the cardiac remodeling mechanisms. Foxp1 downstream target gene transforming growth factor-β1 (TGF-β1) was confirmed by chromatin immunoprecipitation and luciferase assays. Finally, the effects of TGF-β1 blockade on EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes were further confirmed by pharmacological inhibition, more specifically by RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs. RESULTS Foxp1 expression is significantly downregulated in cardiac ECs during angiotensin II-induced cardiac remodeling. EC-Foxp1 deletion results in severe cardiac remodeling, including more cardiac fibrosis with myofibroblast formation and extracellular matrix protein production, as well as decompensated cardiac hypertrophy and further exacerbation of cardiac dysfunction on angiotensin II infusion or transverse aortic constriction operation. In contrast, EC-Foxp1 gain of function protects against pathological cardiac remodeling and improves cardiac dysfunction. TGF-β1 signals are identified as Foxp1 direct target genes, and EC-Foxp1 deletion upregulates TGF-β1 signals to promote myofibroblast formation through fibroblast proliferation and transformation, resulting in severe cardiac fibrosis. Moreover, EC-Foxp1 deletion enhances TGF-β1-promoted endothelin-1 expression, which significantly increases cardiomyocyte size and reactivates cardiac fetal genes, leading to pathological cardiac hypertrophy. Correspondingly, these EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes and cardiac dysfunction are normalized by the blockade of TGF-β1 signals through pharmacological inhibition and RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs. CONCLUSIONS EC-Foxp1 regulates the TGF-β1-endothelin-1 pathway to control pathological cardiac fibrosis and hypertrophy, resulting in cardiac dysfunction. Therefore, targeting the EC-Foxp1-TGF-β1-endothelin-1 pathway might provide a future novel therapy for heart failure.
Collapse
Affiliation(s)
- Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Jingjiang Pi
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Qi Zhang
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Ying Li
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Science, University of Chinese Academy of Sciences (H.W.)
| | - Yajing Shen
- Institute for Biomedical Engineering and Nano Science (Y.S., Y.C.), Tongji University School of Medicine, China
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong (B.T.)
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.)
| | - Zuoren Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Yu Cheng
- Institute for Biomedical Engineering and Nano Science (Y.S., Y.C.), Tongji University School of Medicine, China
| | - Xiangjian Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China (X.Z.).,Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, Australia (X.Z.)
| | - Muredach Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University, New York (M.R.)
| | - Edward Morrisey
- Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, and Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| |
Collapse
|
48
|
Foxp1 is critical for the maintenance of regulatory T-cell homeostasis and suppressive function. PLoS Biol 2019; 17:e3000270. [PMID: 31125332 PMCID: PMC6534289 DOI: 10.1371/journal.pbio.3000270] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/30/2019] [Indexed: 12/13/2022] Open
Abstract
Regulatory T (Treg) cells play central roles in maintaining immune homeostasis and self-tolerance. However, the molecular mechanisms underlying Treg cell homeostasis and suppressive function are still not fully understood. Here, we report that the deletion of another P subfamily members of the forkhead box (Foxp) subfamily member Foxp1 in Treg cells led to increased numbers of activated Treg (aTreg) cells at the expense of quiescent Treg cells, and also resulted in impaired Treg suppressive function. Mice with Foxp1-deficient Treg cells developed spontaneous inflammatory disease with age; they also had more severe inflammatory disease in colitis and experimental autoimmune encephalomyelitis (EAE) models. Mechanistically, we found that Foxp1 bound to the conserved noncoding sequence 2 (CNS2) element of the Foxp3 locus and helped maintain Treg suppressive function by stabilizing the Foxp3 expression. Furthermore, we found that Foxp1 and Foxp3 coordinated the regulation of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression levels. Taken together, our study demonstrates that Foxp1 plays critical roles in both maintaining Treg cell quiescence during homeostasis and regulating Treg suppressive function. The transcription factor Foxp3 has been considered as the master regulator of regulatory T cells (Tregs), but this study shows that another Foxp subfamily member, Foxp1, plays important roles in the homeostasis, stability, and suppressor function of Tregs.
Collapse
|
49
|
Modulation of ADAR mRNA expression in patients with congenital heart defects. PLoS One 2019; 14:e0200968. [PMID: 31039163 PMCID: PMC6490900 DOI: 10.1371/journal.pone.0200968] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 04/05/2019] [Indexed: 12/26/2022] Open
Abstract
Adenosine (A) to inosine (I) RNA editing is a hydrolytic deamination reaction catalyzed by the adenosine deaminase (ADAR) enzyme acting on double-stranded RNA. This posttranscriptional process diversifies a plethora of transcripts, including coding and noncoding RNAs. Interestingly, few studies have been carried out to determine the role of RNA editing in vascular disease. The aim of this study was to determine the potential role of ADARs in congenital heart disease. Strong downregulation of ADAR2 and increase in ADAR1 expression was observed in blood samples from congenital heart disease (CHD) patients. The decrease in expression of ADAR2 was in line with its downregulation in ventricular tissues of dilated cardiomyopathy patients. To further decipher the plausible regulatory pathway of ADAR2 with respect to heart physiology, miRNA profiling of ADAR2 was performed on tissues from ADAR2-/- mouse hearts. Downregulation of miRNAs (miR-29b, miR-405, and miR-19) associated with cardiomyopathy and cardiac fibrosis was observed. Moreover, the upregulation of miR-29b targets COL1A2 and IGF1, indicated that ADAR2 might be involved in cardiac myopathy. The ADAR2 target vascular development associated protein-coding gene filamin B (FLNB) was selected. The editing levels of FLNB were dramatically reduced in ADAR2-/- mice; however, no observable changes in FLNB expression were noted in ADAR2-/- mice compared to wild-type mice. This study proposes that sufficient ADAR2 enzyme activity might play a vital role in preventing cardiovascular defects.
Collapse
|
50
|
Xie Q, Chen C, Li H, Xu J, Wu L, Yu Y, Ren S, Li H, Hua X, Yan H, Rao D, Zhang H, Jin H, Huang H, Huang C. miR-3687 Overexpression Promotes Bladder Cancer Cell Growth by Inhibiting the Negative Effect of FOXP1 on Cyclin E2 Transcription. Mol Ther 2019; 27:1028-1038. [PMID: 30935821 DOI: 10.1016/j.ymthe.2019.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/07/2019] [Accepted: 03/01/2019] [Indexed: 12/17/2022] Open
Abstract
Cyclin E2, a member of the cyclin family, is a key cell cycle-related protein. This protein plays essential roles in cancer progression, and, as such, an inhibitor of cyclin E2 has been approved to treat several types of cancers. Even so, mechanisms underlying how to regulate cyclin E2 expression in cancer remain largely unknown. In the current study, miR-3687 was upregulated in clinical bladder cancer (BC) tumor tissues, The Cancer Genome Atlas (TCGA) database, and human BC cell lines. Inhibition of miR-3687 expression significantly reduced human BC cell proliferation in vitro and tumor growth in vivo, which coincided with the induction of G0/G1 cell cycle arrest and downregulation of cyclin E2 protein expression. Interestingly, overexpression of cyclin E2 reversed the inhibition of BC proliferation induced by miR-3687. Mechanistic studies suggested that miR-3687 binds to the 3' UTR of foxp1 mRNA, downregulates FOXP1 protein expression, and in turn promotes the transcription of cyclin E2, thereby promoting the growth of BC cells. Collectively, the current study not only establishes a novel regulatory axis of miR-3687/FOXP1 regarding regulation of cyclin E2 expression in BC cells, but also provides strong suggestive evidence that miR-3687 and FOXP1 may be promising targets in therapeutic strategies for human BC.
Collapse
Affiliation(s)
- Qipeng Xie
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Caiyi Chen
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haiying Li
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiheng Xu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lei Wu
- Heze Municipal Hospital, Heze, Shandong, 274031, China
| | - Yuan Yu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shuwei Ren
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyan Li
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaohui Hua
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huiying Yan
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Dapang Rao
- The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huxiang Zhang
- Biobank of Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Chuanshu Huang
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10010, USA.
| |
Collapse
|