1
|
Japa O, Phuangsri C, Klinbumrung K, Prakhammin K. Receptor-regulated smads (R-Smads) in the liver fluke Fasciola gigantica: Characterization, comparative sequence analysis, and life stage-specific expression. Acta Trop 2025; 264:107603. [PMID: 40157585 DOI: 10.1016/j.actatropica.2025.107603] [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: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The transforming growth factor-beta (TGF-β) signaling pathway is a key conserved developmental pathway, demonstrating remarkable conservation in its components, structures, and functions across diverse animal species. This study presents a comprehensive characterization and comparative analysis of receptor-regulated Smads (R-Smads) in Fasciola gigantica, a major parasite affecting cattle, buffalo, and ruminants in tropical regions. Four R-Smads were identified: FgSmad1/5, FgSmad2, FgSmad3, and FgSmad8. Full-length cDNAs for these R-Smads, isolated using the rapid amplification of cDNA ends (RACE) approach, were 3348, 3010, 5172, and 1991 bp in length, encoding proteins of 464, 506, 678, and 404 amino acids, respectively. FgSmad1/5 and FgSmad8 were classified as bone morphogenetic protein (BMP)-specific R-Smads (BR-Smads) due to their conserved signature motifs and similarity to those found in parasitic platyhelminths and mammalian hosts. Conversely, FgSmad2 and FgSmad3 were identified as TGF-β/activin-specific R-Smads (AR-Smads), displaying a distinctive pattern of conserved motifs commonly observed among trematode parasites. Notably, FgSmad2 exhibited an atypical feature for R-Smads, as it lacked the MH1 domain. The expressions of F. gigantica R-Smad genes were observed across all developmental stages, with the highest levels of most R-Smads occurring in the unembryonated egg stage (primarily BR-Smads) and the metacercaria stage (predominantly AR-Smads), underscoring a significant shift in TGF-β and BMP signaling dynamics during development. These findings greatly advance our understanding of R-Smad proteins within the TGF-β signaling pathway of F. gigantica and provide a solid foundation for future research into their roles and regulatory mechanisms.
Collapse
Affiliation(s)
- Ornampai Japa
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao, Thailand; Scientific Instrument and Product Standard Quality Inspection Center, University of Phayao, Phayao, Thailand.
| | - Chorpaka Phuangsri
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Khuruwan Klinbumrung
- Scientific Instrument and Product Standard Quality Inspection Center, University of Phayao, Phayao, Thailand
| | - Khanuengnij Prakhammin
- Department of Applied Statistics, Rajamangala University of Technology Isan, Khon Kaen Campus, Khon Kaen, Thailand
| |
Collapse
|
2
|
Bakheet T, Al-Mutairi N, Doubi M, Al-Ahmadi W, Alhosaini K, Al-Zoghaibi F. A Computational Recognition Analysis of Promising Prognostic Biomarkers in Breast, Colon and Lung Cancer Patients. Int J Mol Sci 2025; 26:1017. [PMID: 39940786 PMCID: PMC11817791 DOI: 10.3390/ijms26031017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Breast, colon, and lung carcinomas are classified as aggressive tumors with poor relapse-free survival (RFS), progression-free survival (PF), and poor hazard ratios (HRs) despite extensive therapy. Therefore, it is essential to identify a gene expression signature that correlates with RFS/PF and HR status in order to predict treatment efficiency. RNA-binding proteins (RBPs) play critical roles in RNA metabolism, including RNA transcription, maturation, and post-translational regulation. However, their involvement in cancer is not yet fully understood. In this study, we used computational bioinformatics to classify the functions and correlations of RBPs in solid cancers. We aimed to identify molecular biomarkers that could help predict disease prognosis and improve the therapeutic efficiency in treated patients. Intersection analysis summarized more than 1659 RBPs across three recently updated RNA databases. Bioinformatics analysis showed that 58 RBPs were common in breast, colon, and lung cancers, with HR values < 1 and >1 and a significant Q-value < 0.0001. RBP gene clusters were identified based on RFS/PF, HR, p-value, and fold induction. To define union RBPs, common genes were subjected to hierarchical clustering and were classified into two groups. Poor survival was associated with high genes expression, including CDKN2A, MEX3A, RPL39L, VARS, GSPT1, SNRPE, SSR1, and TIA1 in breast and colon cancer but not with lung cancer; and poor survival was associated with low genes expression, including PPARGC1B, EIF4E3, and SMAD9 in breast, colon, and lung cancer. This study highlights the significant contribution of PPARGC1B, EIF4E3, and SMAD9 out of 11 RBP genes as prognostic predictors in patients with breast, colon, and lung cancers and their potential application in personalized therapy.
Collapse
Affiliation(s)
- Tala Bakheet
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (T.B.); (N.A.-M.); (M.D.); (W.A.-A.)
| | - Nada Al-Mutairi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (T.B.); (N.A.-M.); (M.D.); (W.A.-A.)
| | - Mosaab Doubi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (T.B.); (N.A.-M.); (M.D.); (W.A.-A.)
| | - Wijdan Al-Ahmadi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (T.B.); (N.A.-M.); (M.D.); (W.A.-A.)
| | - Khaled Alhosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Fahad Al-Zoghaibi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (T.B.); (N.A.-M.); (M.D.); (W.A.-A.)
| |
Collapse
|
3
|
Shweta, Sharma K, Shakarad M, Agrawal N, Maurya SK. Drosophila glial system: an approach towards understanding molecular complexity of neurodegenerative diseases. Mol Biol Rep 2024; 51:1146. [PMID: 39532789 DOI: 10.1007/s11033-024-10075-w] [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: 09/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Glia is pivotal in regulating neuronal stem cell proliferation, functioning, and nervous system homeostasis, significantly influencing neuronal health and disorders. Dysfunction in glial activity is a key factor in the development and progression of brain pathology. However, a deeper understanding of the intricate nature of glial cells and their diverse role in neurological disorders is still required. To this end, we conducted data mining to retrieve literature from PubMed and Google Scholar using the keywords: glia, Drosophila, neurodegeneration, and mammals. The retrieved literature was manually screened and used to comprehensively understand and present the different glial types in Drosophila, i.e., perineurial, subperineurial, cortex, astrocyte-like and ensheathing glia, their relevance with mammalian counterparts, mainly microglia and astrocytes, and their potential to reveal complex neuron-glial molecular networks in managing neurodegenerative processes.
Collapse
Affiliation(s)
- Shweta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Khushboo Sharma
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallikarjun Shakarad
- Evolutionary Biology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Namita Agrawal
- Fly Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
4
|
Songkoomkrong S, Nonkhwao S, Duangprom S, Saetan J, Manochantr S, Sobhon P, Kornthong N, Amonruttanapun P. Investigating the potential effect of Holothuria scabra extract on osteogenic differentiation in preosteoblast MC3T3-E1 cells. Sci Rep 2024; 14:26415. [PMID: 39488645 PMCID: PMC11531581 DOI: 10.1038/s41598-024-77850-4] [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: 07/20/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
The present medical treatments of osteoporosis come with adverse effects. It leads to the exploration of natural products as safer alternative medical prevention and treatment. The sea cucumber, Holothuria scabra, has commercial significance in Asian countries with rising awareness of its properties as a functional food. This study aims to investigate the effects of the inner wall (IW) extract isolated from H. scabra on extracellular matrix maturation, mineralization, and osteogenic signaling pathways on MC3T3-E1 preosteoblasts. The IW showed the expression of several growth factors. Molecular docking revealed that H. scabra BMP2/4 binds specifically to mammal BMP2 type I receptor (BMPR-IA). After osteogenic induction, the viability of cells treated with IW extract was assessed and designated with treatment of 0.1, 0.5, 1, and 5 µg/ml of IW extract for 21 consecutive days. On days 14 and 21, treatments with IW extract at 1 and 5 µg/ml showed increased alkaline phosphatase (ALP) activity and calcium deposit levels in a dose-dependent manner compared to the control group. Moreover, the transcriptomic analysis of total RNA of cells treated with 5 µg/ml of IW extract exhibited upregulation of TGF-β, PI3K/Akt, MAPK, Wnt and PTH signaling pathways at days 14. This study suggests that IW extract from H. scabra exhibits the potential to enhance osteogenic differentiation and mineralization of MC3T3-E1 preosteoblasts through TGF-β, PI3K/Akt, MAPK, Wnt and PTH signaling pathways. Further investigation into the molecular mechanisms underlying the effect of IW extract on osteogenesis is crucial to support its application as a naturally derived supplement for prevention or treatment of osteoporosis.
Collapse
Affiliation(s)
- Sineenart Songkoomkrong
- Chulabhorn International College of Medicine, Thammasat University, Rangsit campus, Pathumthani, 12121, Thailand
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12121, Thailand
| | - Siriporn Nonkhwao
- Chulabhorn International College of Medicine, Thammasat University, Rangsit campus, Pathumthani, 12121, Thailand
| | - Supawadee Duangprom
- Chulabhorn International College of Medicine, Thammasat University, Rangsit campus, Pathumthani, 12121, Thailand
| | - Jirawat Saetan
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
| | - Sirikul Manochantr
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12121, Thailand
- Center of Excellence in Stem Cell Research and Innovation, Thammasat University, Pathumthani, 12121, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Napamanee Kornthong
- Chulabhorn International College of Medicine, Thammasat University, Rangsit campus, Pathumthani, 12121, Thailand
| | - Prateep Amonruttanapun
- Chulabhorn International College of Medicine, Thammasat University, Rangsit campus, Pathumthani, 12121, Thailand.
| |
Collapse
|
5
|
Zhao J, Tang X, Zhu H. Chondroitin polymerizing factor (CHPF) promotes the progression of colorectal cancer through ASB2-mediated ubiquitylation of SMAD9. Histol Histopathol 2024; 39:1493-1503. [PMID: 38591191 DOI: 10.14670/hh-18-738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Chondroitin polymerizing factor (CHPF) has been reported to play a pivotal role in the progression of multiple cancers, however, the relationship between CHPF and colorectal cancer (CRC) progression has not been fully understood. The current study revealed that CHPF expression was upregulated in patients with CRC and correlated with an unfavorable prognosis. Also, CHPF knockdown effectively suppressed the viability and mobility of CRC cells and the growth of xenograft tumors. Additionally, SMAD9 was identified as a downstream target of CHPF. SMAD9 knockdown successfully abrogated the promotion of CHPF overexpression in CRC progression, indicating that CHPF regulated the development of CRC through SMAD9. Mechanistically, SMAD9 is ubiquitinated by ASB2, and the regulatory effect of CHPF on SMAD9 activity was exerted via its mediation of ASB2. Collectively, CHPF functioned as a promising prognostic biomarker and tumor-promoter of CRC by regulating the ASB2-mediated ubiquitination of SMAD9.
Collapse
Affiliation(s)
- Jiang Zhao
- Department of Oncology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, PR China
| | - Xiaolong Tang
- Department of Oncology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, PR China
| | - Huijun Zhu
- Department of Traditional Chinese Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, PR China.
| |
Collapse
|
6
|
Gençel D, Erbil NN, Demiryürek Ş, Demiryürek AT. Current and emerging treatment modalities for fibrodysplasia ossificans progressiva. Expert Opin Pharmacother 2024; 25:2225-2234. [PMID: 39451784 DOI: 10.1080/14656566.2024.2422548] [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: 07/27/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Heterotopic ossification (HO), acquired or hereditary, is a diverse pathological condition defined by the production of extraskeletal bone in muscles, soft tissues, and connective tissues. Acquired HO is relatively prevalent and develops mostly in response to trauma, although its etiology is unknown. Genetic forms provide insight into the pathobiological mechanisms of this disorder. Fibrodysplasia ossificans progressiva (FOP) is a rare hereditary form of HO that can have a significant impact on affected individuals. FOP steadily weakens affected subjects and increases their risk of death. AREAS COVERED The U.S. Food and Drug Administration has recently approved the retinoid palovarotene as the first compound to treat heterotopic ossification in patients with FOP. This review provides a comprehensive overview of current and potential future pharmacotherapeutic options and their modes of action. The online databases PubMed, Cochrane Library, Web of Science, and ClinicalTrials.gov were searched using the terms 'heterotopic ossification' and 'fibrodysplasia ossificans progressiva' or synonyms, with a special focus over the last 5 years of publications. EXPERT OPINION Approval of palovarotene, as the first retinoid indicated for reduction in the volume of new HO, may revolutionize the therapeutic landscape. However, long-term safety and efficacy data for palovarotene are currently lacking.
Collapse
Affiliation(s)
- Dilan Gençel
- Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Nejla Nur Erbil
- Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Şeniz Demiryürek
- Department of Physiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | | |
Collapse
|
7
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
8
|
Li X, Liu L, Wan MX, Gong LM, Su J, Xu L. Active Components of Pueraria lobata through the MAPK/ERK Signaling Pathway Alleviate Iron Overload in Alcoholic Liver Disease. Chem Biodivers 2024; 21:e202400005. [PMID: 38504590 DOI: 10.1002/cbdv.202400005] [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: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE To delve into the primary active ingredients and mechanism of Pueraria lobata for alleviating iron overload in alcoholic liver disease. METHODS Pueraria lobata's potential targets and signaling pathways in treating alcohol-induced iron overloads were predicted using network pharmacology analysis. Then, animal experiments were used to validate the predictions of network pharmacology. The impact of puerarin or genistein on alcohol-induced iron accumulation, liver injury, oxidative stress, and apoptosis was assessed using morphological examination, biochemical index test, and immunofluorescence. Key proteins implicated in linked pathways were identified using RT-qPCR, western blot analysis, and immunohistochemistry. RESULTS Network pharmacological predictions combined with animal experiments suggest that the model group compared to the control group, exhibited activation of the MAPK/ERK signaling pathway, suppression of hepcidin expression, and aggravated iron overload, liver damage, oxidative stress, and hepatocyte death. Puerarin and genistein, the active compounds in Pueraria lobata, effectively mitigated the aforementioned alcohol-induced effects. No statistically significant disparities were seen in the effects above between the two groups receiving drug therapy. CONCLUSION This study preliminarily demonstrated that puerarin and genistein in Pueraria lobata may increase hepcidin production to alleviate alcohol-induced iron overload by inhibiting the MAPK/ERK signaling pathway.
Collapse
Affiliation(s)
- Xue Li
- College of Basic Medicine, Dali University, Dali, China
| | - Le Liu
- College of Basic Medicine, Dali University, Dali, China
| | - Mei-Xuan Wan
- College of Basic Medicine, Dali University, Dali, China
| | - Li-Min Gong
- College of Basic Medicine, Dali University, Dali, China
| | - Juan Su
- College of Basic Medicine, Dali University, Dali, China
| | - Li Xu
- College of Basic Medicine, Dali University, Dali, China
- Yunnan Key Laboratory of Screening and Research on Anti-pathogenic Plant Resources from Western Yunnan, Dali University, Dali, China
| |
Collapse
|
9
|
Thériault S, Li Z, Abner E, Luan J, Manikpurage HD, Houessou U, Zamani P, Briend M, Boudreau DK, Gaudreault N, Frenette L, Argaud D, Dahmene M, Dagenais F, Clavel MA, Pibarot P, Arsenault BJ, Boekholdt SM, Wareham NJ, Esko T, Mathieu P, Bossé Y. Integrative genomic analyses identify candidate causal genes for calcific aortic valve stenosis involving tissue-specific regulation. Nat Commun 2024; 15:2407. [PMID: 38494474 PMCID: PMC10944835 DOI: 10.1038/s41467-024-46639-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
There is currently no medical therapy to prevent calcific aortic valve stenosis (CAVS). Multi-omics approaches could lead to the identification of novel molecular targets. Here, we perform a genome-wide association study (GWAS) meta-analysis including 14,819 cases among 941,863 participants of European ancestry. We report 32 genomic loci, among which 20 are novel. RNA sequencing of 500 human aortic valves highlights an enrichment in expression regulation at these loci and prioritizes candidate causal genes. Homozygous genotype for a risk variant near TWIST1, a gene involved in endothelial-mesenchymal transition, has a profound impact on aortic valve transcriptomics. We identify five genes outside of GWAS loci by combining a transcriptome-wide association study, colocalization, and Mendelian randomization analyses. Using cross-phenotype and phenome-wide approaches, we highlight the role of circulating lipoproteins, blood pressure and inflammation in the disease process. Our findings pave the way for the development of novel therapies for CAVS.
Collapse
Affiliation(s)
- Sébastien Thériault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada.
| | - Zhonglin Li
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Hasanga D Manikpurage
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Ursula Houessou
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Pardis Zamani
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Mewen Briend
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Dominique K Boudreau
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Lily Frenette
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Déborah Argaud
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Manel Dahmene
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - François Dagenais
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Pibarot
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Benoit J Arsenault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Patrick Mathieu
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
10
|
Wang C, Liu Z, Zeng Y, Zhou L, Long Q, Hassan IU, Zhang Y, Qi X, Cai D, Mao B, Lu G, Sun J, Yao Y, Deng Y, Zhao Q, Feng B, Zhou Q, Chan WY, Zhao H. ZSWIM4 regulates embryonic patterning and BMP signaling by promoting nuclear Smad1 degradation. EMBO Rep 2024; 25:646-671. [PMID: 38177922 PMCID: PMC10897318 DOI: 10.1038/s44319-023-00046-w] [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: 02/23/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024] Open
Abstract
The dorsoventral gradient of BMP signaling plays an essential role in embryonic patterning. Zinc Finger SWIM-Type Containing 4 (zswim4) is expressed in the Spemann-Mangold organizer at the onset of Xenopus gastrulation and is then enriched in the developing neuroectoderm at the mid-gastrula stages. Knockdown or knockout of zswim4 causes ventralization. Overexpression of zswim4 decreases, whereas knockdown of zswim4 increases the expression levels of ventrolateral mesoderm marker genes. Mechanistically, ZSWIM4 attenuates the BMP signal by reducing the protein stability of SMAD1 in the nucleus. Stable isotope labeling by amino acids in cell culture (SILAC) identifies Elongin B (ELOB) and Elongin C (ELOC) as the interaction partners of ZSWIM4. Accordingly, ZSWIM4 forms a complex with the Cul2-RING ubiquitin ligase and ELOB and ELOC, promoting the ubiquitination and degradation of SMAD1 in the nucleus. Our study identifies a novel mechanism that restricts BMP signaling in the nucleus.
Collapse
Affiliation(s)
- Chengdong Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziran Liu
- Qingdao Municipal Center for Disease Control and Prevention, 266033, Qingdao, Shandong, China
| | - Yelin Zeng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liangji Zhou
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Long
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Imtiaz Ul Hassan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuanliang Zhang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xufeng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Dongqing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Bingyu Mao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Chinese Academy of Sciences, Kunming, China
| | - Gang Lu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jianmin Sun
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, No. 1160 Shengli Street, 750004, Yinchuan, China
| | - Yonggang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Chinese Academy of Sciences, Kunming, China
| | - Yi Deng
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Bo Feng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qin Zhou
- School of Basic Medical Sciences, Harbin Medical University, 150081, Harbin, China
| | - Wai Yee Chan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
11
|
Zhao H, Duan R, Wang Q, Hu X, Zhao Q, Wu W, Jiang R, Gong S, Wang L, Liu J, Deng J, Liang H, Miao Y, Yuan P. MiR-122-5p as a potential regulator of pulmonary vascular wall cell in idiopathic pulmonary arterial hypertension. Heliyon 2023; 9:e22922. [PMID: 38144299 PMCID: PMC10746431 DOI: 10.1016/j.heliyon.2023.e22922] [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: 11/21/2022] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
MicroRNAs (miRNAs) are versatile regulators of pulmonary arterial remodeling in idiopathic pulmonary arterial hypertension (IPAH). We herein aimed to characterize miRNAs in peripheral blood mononuclear cell (PBMC) and plasma exosomes, and investigate specific miRNA expression in pulmonary artery cells and lung tissues in IPAH. A co-dysregulated miRNA was identified from the miRNA expression profiles of PBMC and plasma exosomes in IPAH. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed the potential function of differentially expressed miRNAs. Real-time quantitative reverse transcription polymerase chain reaction was used to validate the expression of specific miRNAs in hypoxia-induced pulmonary microvascular endothelial cells (PMECs), pulmonary artery smooth muscle cells (PASMCs), pericyte cells (PCs), and lung tissues of patients with IPAH and rats. Finally, the miRNA-mRNA mechanisms of miR-122-5p were predicted. MiR-122-5p was the only co-upregulated miRNA in PBMC and plasma exosomes in patients with IPAH. Functional analysis of differentially expressed miRNAs revealed associations with the GO terms "transcription, DNA-templated," "cytoplasm," and "metal ion binding" in both PBMC and plasma exosomes, KEGG pathway MAPK signaling in PBMC, and KEGG-pathway human papillomavirus infection in plasma exosomes. Hypoxic PMECs and PCs, lung tissue of patients with IPAH, and rats showed increased expression of miR-122-5p, but hypoxic PASMCs showed decreased expression. And miR-122-5p mimics and inhibitor affected cell proliferation. Finally, miR-122-5p was found to potentially target DLAT (in lung tissue) and RIMS1 (in PMECs) in IPAH. According to the dual-luciferase assay, miR-122-5p bound to DLAT or RIMS1. In studies, DLAT imbalance was associated with cell proliferation and migration, RIMS1 is differentially expressed in cancer and correlated with cancer prognosis. Our findings suggest that the miR-122-5p is involved in various biological functions in the adjacent vascular wall cells in IPAH.
Collapse
Affiliation(s)
- Hui Zhao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai 200093, China
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ruowang Duan
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Qian Wang
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai 200093, China
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Xiaoyi Hu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Qinhua Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Wenhui Wu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Sugang Gong
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jinming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jie Deng
- Southern Medical University, Guangzhou, 510000, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou, Jiangsu Province, 215125, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
12
|
Ahsan T, Shoily SS, Ahmed T, Sajib AA. Role of the redox state of the Pirin-bound cofactor on interaction with the master regulators of inflammation and other pathways. PLoS One 2023; 18:e0289158. [PMID: 38033031 PMCID: PMC10688961 DOI: 10.1371/journal.pone.0289158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/10/2023] [Indexed: 12/02/2023] Open
Abstract
Persistent cellular stress induced perpetuation and uncontrolled amplification of inflammatory response results in a shift from tissue repair toward collateral damage, significant alterations of tissue functions, and derangements of homeostasis which in turn can lead to a large number of acute and chronic pathological conditions, such as chronic heart failure, atherosclerosis, myocardial infarction, neurodegenerative diseases, diabetes, rheumatoid arthritis, and cancer. Keeping the vital role of balanced inflammation in maintaining tissue integrity in mind, the way to combating inflammatory diseases may be through identification and characterization of mediators of inflammation that can be targeted without hampering normal body function. Pirin (PIR) is a non-heme iron containing protein having two different conformations depending on the oxidation state of the iron. Through exploration of the Pirin interactome and using molecular docking approaches, we identified that the Fe2+-bound Pirin directly interacts with BCL3, NFKBIA, NFIX and SMAD9 with more resemblance to the native binding pose and higher affinity than the Fe3+-bound form. In addition, Pirin appears to have a function in the regulation of inflammation, the transition between the canonical and non-canonical NF-κB pathways, and the remodeling of the actin cytoskeleton. Moreover, Pirin signaling appears to have a critical role in tumor invasion and metastasis, as well as metabolic and neuro-pathological complications. There are regulatory variants in PIR that can influence expression of not only PIR but also other genes, including VEGFD and ACE2. Disparity exists between South Asian and European populations in the frequencies of variant alleles at some of these regulatory loci that may lead to differential occurrence of Pirin-mediated pathogenic conditions.
Collapse
Affiliation(s)
- Tamim Ahsan
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka, Bangladesh
| | - Sabrina Samad Shoily
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Tasnim Ahmed
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
13
|
Wu Y, Liang H, Luo A, Li Y, Liu Z, Li X, Li W, Liang K, Li J, Liu Z, Du Y. Gelatin-based 3D biomimetic scaffolds platform potentiates culture of cancer stem cells in esophageal squamous cell carcinoma. Biomaterials 2023; 302:122323. [PMID: 37717405 DOI: 10.1016/j.biomaterials.2023.122323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/02/2023] [Accepted: 09/09/2023] [Indexed: 09/19/2023]
Abstract
Cancer stem cells (CSCs) are crucial for tumorigenesis, metastasis, and therapy resistance in esophageal squamous cell carcinoma (ESCC). To further elucidate the mechanism underlying characteristics of CSCs and develop CSCs-targeted therapy, an efficient culture system that could expand and maintain CSCs is needed. CSCs reside in a complex tumor microenvironment, and three-dimensional (3D) culture systems of biomimetic scaffolds are expected to better support the growth of CSCs by recapitulating the biophysical properties of the extracellular matrix (ECM). Here, we established gelatin-based 3D biomimetic scaffolds mimicking the stiffness and collagen content of ESCC, which could enrich ESCC CSCs efficiently. Biological changes of ESCC cells laden in scaffolds with three different viscoelasticity emulating physiological stiffness of esophageal tissues were thoroughly investigated in varied aspects such as cell morphology, viability, cell phenotype markers, and transcriptomic profiling. The results demonstrated the priming effects of viscoelasticity on the stemness of ESCC. The highly viscous scaffolds (G': 6-403 Pa; G'': 2-75 Pa) better supported the enrichment of ESCC CSCs, and the TGF-beta signaling pathway might be involved in regulating the stemness of ESCC cells. Compared to two-dimensional (2D) cultures, highly viscous scaffolds significantly promoted the clonal expansion of ESCC cells in vitro and tumor formation ability in vivo. Our findings highlight the crucial role of biomaterials' viscoelasticity for the 3D culture of ESCC CSCs in vitro, and this newly-established culture system represents a valuable platform to support their growth, which could facilitate the CSCs-targeted therapy in the future.
Collapse
Affiliation(s)
- Yenan Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haiwei Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Aiping Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiqiang Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenxin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
14
|
Hashimoto K, Miyagawa Y, Watanabe S, Takasaki K, Nishizawa M, Yatsuki K, Takahashi Y, Kamata H, Kihira C, Hiraike H, Sasamori Y, Kido K, Ryo E, Nagasaka K. The TGF-β/UCHL5/Smad2 Axis Contributes to the Pathogenesis of Placenta Accreta. Int J Mol Sci 2023; 24:13706. [PMID: 37762005 PMCID: PMC10530686 DOI: 10.3390/ijms241813706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Placenta accreta is a high-risk condition causing obstetric crisis and hemorrhage; however, its pathogenesis remains unknown. We aimed to identify the factors contributing to trophoblast invasiveness and angiogenic potential, which in turn drive the pathogenesis of placenta accreta. We focused on the transforming growth factor (TGF)-β1-Smad pathway and investigated the intrinsic relationship between the time- and dose-dependent inhibition of the ubiquitinating enzyme UCHL5 using bAP15, a deubiquitinase inhibitor, after TGF-β1 stimulation and the invasive and angiogenic potential of two cell lines, gestational choriocarcinoma cell line JEG-3 and trophoblast cell line HTR-8/SVneo. UCHL5 inhibition negatively regulated TGF-β1-induced Smad2 activation, decreasing extravillous trophoblast invasiveness. Smad1/5/9 and extracellular signal-regulated kinase (ERK) were simultaneously activated, and vascular endothelial growth factor was secreted into the trophoblast medium. However, extravillous trophoblast culture supernatant severely impaired the vasculogenic potential of human umbilical vein endothelial cells. These results suggest that the downstream ERK pathway and Smad1/5/9 potentially regulate the TGF-β1-Smad pathway in extravillous trophoblasts, whereas Smad2 contributes to their invasiveness. The abnormal invasive and angiogenic capacities of extravillous cells, likely driven by the interaction between TGF-β1-Smad and ERK pathways, underlie the pathogenesis of placenta accreta.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kazunori Nagasaka
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| |
Collapse
|
15
|
Wang X, Mei D, Lu Z, Zhang Y, Sun Y, Lu T, Yan H, Yue W. Genome-wide association study identified six loci associated with adverse drug reactions to aripiprazole in schizophrenia patients. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:44. [PMID: 37491364 PMCID: PMC10368716 DOI: 10.1038/s41537-023-00369-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023]
Abstract
Aripiprazole is recommended for routine use in schizophrenia patients. However, the biological mechanism for the adverse drug reactions (ADRs) among schizophrenia patients with the antipsychotic drug aripiprazole is far from clear. To explore the potential genetic factors that may cause movement-related adverse antipsychotic effects in patients, we conducted an association analysis between movement-related ADRs and SNPs in schizophrenia patients receiving aripiprazole monotherapy. In this study, multiple ADRs of 384 patients were quantified within 6-week treatment, and the scores of movement-related ADRs at baseline and follow-up time points during treatment were obtained. The highest score record was used as the quantitative index in analysis, and genetic analysis at the genome-wide level was conducted. The SNP rs4149181 in SLC22A8 [P = 2.28 × 10-8] showed genome-wide significance, and rs2284223 in ADCYAP1R1 [P = 9.76 × 10-8], rs73258503 in KCNIP4 [P = 1.39 × 10-7], rs678428 in SMAD9 [P = 4.70 × 10-7], rs6421034 in NAP1L4 [P = 6.80 × 10-7], and rs1394796 in ERBB4 [P = 8.60 × 10-7] were found to be significantly associated with movement-related ADRs. The combined prediction model of these six loci showed acceptable performance in predicting adverse events [area under the curve (AUC): 0.84]. Combined with the function and network of the above genes and other candidate loci (KCNA1, CACNG1, etc.), we hypothesize that SLC22A8 and KCNIP4-Kv channel perform their respective functions as transporter or channel and participate in the in vivo metabolism or effects of aripiprazole. The above results imply the important function of ion transporters and channels in movement-related adverse antipsychotic effects in aripiprazole monotherapy schizophrenia patients.
Collapse
Affiliation(s)
- Xueping Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China
| | - Dongli Mei
- School of Nursing, Peking University, 10019, Beijing, China
| | - Zhe Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China
| | - Yuyanan Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China
| | - Yaoyao Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China
| | - Tianlan Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China
| | - Hao Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China.
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), 100191, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.
| |
Collapse
|
16
|
Katagiri T, Tsukamoto S, Kuratani M, Tsuji S, Nakamura K, Ohte S, Kawaguchi Y, Takaishi K. A blocking monoclonal antibody reveals dimerization of intracellular domains of ALK2 associated with genetic disorders. Nat Commun 2023; 14:2960. [PMID: 37231012 PMCID: PMC10212922 DOI: 10.1038/s41467-023-38746-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Mutations in activin receptor-like kinase 2 (ALK2) can cause the pathological osteogenic signaling seen in some patients with fibrodysplasia ossificans progressiva and other conditions such as diffuse intrinsic pontine glioma. Here, we report that intracellular domain of wild-type ALK2 readily dimerizes in response to BMP7 binding to drive osteogenic signaling. This osteogenic signaling is pathologically triggered by heterotetramers of type II receptor kinases and ALK2 mutant forms, which form intracellular domain dimers in response to activin A binding. We develop a blocking monoclonal antibody, Rm0443, that can suppress ALK2 signaling. We solve the crystal structure of the ALK2 extracellular domain complex with a Fab fragment of Rm0443 and show that Rm0443 induces dimerization of ALK2 extracellular domains in a back-to-back orientation on the cell membrane by binding the residues H64 and F63 on opposite faces of the ligand-binding site. Rm0443 could prevent heterotopic ossification in a mouse model of fibrodysplasia ossificans progressiva that carries the human R206H pathogenic mutant.
Collapse
Affiliation(s)
- Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
| | - Sho Tsukamoto
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Mai Kuratani
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Shinnosuke Tsuji
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Satoshi Ohte
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yoshiro Kawaguchi
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kiyosumi Takaishi
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
17
|
Upton PD, Dunmore BJ, Li W, Morrell NW. An emerging class of new therapeutics targeting TGF, Activin, and BMP ligands in pulmonary arterial hypertension. Dev Dyn 2023; 252:327-342. [PMID: 35434863 PMCID: PMC10952790 DOI: 10.1002/dvdy.478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an often fatal condition, the primary pathology of which involves loss of pulmonary vascular perfusion due to progressive aberrant vessel remodeling. The reduced capacity of the pulmonary circulation places increasing strain on the right ventricle of the heart, leading to death by heart failure. Currently, licensed therapies are primarily vasodilators, which have increased the median post-diagnosis life expectancy from 2.8 to 7 years. Although this represents a substantial improvement, the search continues for transformative therapeutics that reverse established disease. The genetics of human PAH heavily implicates reduced endothelial bone morphogenetic protein (BMP) signaling as a causal role for the disease pathobiology. Recent approaches have focused on directly enhancing BMP signaling or removing the inhibitory influence of pathways that repress BMP signaling. In this critical commentary, we review the evidence underpinning the development of two approaches: BMP-based agonists and inhibition of activin/GDF signaling. We also address the key considerations and questions that remain regarding these approaches.
Collapse
Affiliation(s)
- Paul D. Upton
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Benjamin J. Dunmore
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Wei Li
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Nicholas W. Morrell
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| |
Collapse
|
18
|
The study of selection signature and its applications on identification of candidate genes using whole genome sequencing data in chicken - a review. Poult Sci 2023; 102:102657. [PMID: 37054499 PMCID: PMC10123265 DOI: 10.1016/j.psj.2023.102657] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Chicken is a major source of protein for the increasing human population and is useful for research purposes. There are almost 1,600 distinct regional breeds of chicken across the globe, among which a large body of genetic and phenotypic variations has been accumulated due to extensive natural and artificial selection. Moreover, natural selection is a crucial force for animal domestication. Several approaches have been adopted to detect selection signatures in different breeds of chicken using whole genome sequencing (WGS) data including integrated haplotype score (iHS), cross-populated extend haplotype homozygosity test (XP-EHH), fixation index (FST), cross-population composite likelihood ratio (XP-CLR), nucleotide diversity (Pi), and others. In addition, gene enrichment analyses are utilized to determine KEGG pathways and gene ontology (GO) terms related to traits of interest in chicken. Herein, we review different studies that have adopted diverse approaches to detect selection signatures in different breeds of chicken. This review systematically summarizes different findings on selection signatures and related candidate genes in chickens. Future studies could combine different selection signatures approaches to strengthen the quality of the results thereby providing more affirmative inference. This would further aid in deciphering the importance of selection in chicken conservation for the increasing human population.
Collapse
|
19
|
Bergen DJM, Maurizi A, Formosa MM, McDonald GLK, El-Gazzar A, Hassan N, Brandi ML, Riancho JA, Rivadeneira F, Ntzani E, Duncan EL, Gregson CL, Kiel DP, Zillikens MC, Sangiorgi L, Högler W, Duran I, Mäkitie O, Van Hul W, Hendrickx G. High Bone Mass Disorders: New Insights From Connecting the Clinic and the Bench. J Bone Miner Res 2023; 38:229-247. [PMID: 36161343 PMCID: PMC10092806 DOI: 10.1002/jbmr.4715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
Monogenic high bone mass (HBM) disorders are characterized by an increased amount of bone in general, or at specific sites in the skeleton. Here, we describe 59 HBM disorders with 50 known disease-causing genes from the literature, and we provide an overview of the signaling pathways and mechanisms involved in the pathogenesis of these disorders. Based on this, we classify the known HBM genes into HBM (sub)groups according to uniform Gene Ontology (GO) terminology. This classification system may aid in hypothesis generation, for both wet lab experimental design and clinical genetic screening strategies. We discuss how functional genomics can shape discovery of novel HBM genes and/or mechanisms in the future, through implementation of omics assessments in existing and future model systems. Finally, we address strategies to improve gene identification in unsolved HBM cases and highlight the importance for cross-laboratory collaborations encompassing multidisciplinary efforts to transfer knowledge generated at the bench to the clinic. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Dylan J M Bergen
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK.,Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Melissa M Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta.,Center for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Georgina L K McDonald
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Ahmed El-Gazzar
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Neelam Hassan
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | | | - José A Riancho
- Department of Internal Medicine, Hospital U M Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece.,Center for Evidence Synthesis in Health, Policy and Practice, Center for Research Synthesis in Health, School of Public Health, Brown University, Providence, RI, USA.,Institute of Biosciences, University Research Center of loannina, University of Ioannina, Ioannina, Greece
| | - Emma L Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Department of Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Celia L Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Douglas P Kiel
- Marcus Institute for Aging Research, Hebrew SeniorLife and Department of Medicine Beth Israel Deaconess Medical Center and Harvard Medical School, Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Luca Sangiorgi
- Department of Rare Skeletal Diseases, IRCCS Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | | | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Centre, Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
20
|
Lopez MA, Si Y, Hu X, Williams V, Qushair F, Carlyle J, Alesce L, Conklin M, Gilbert S, Bamman MM, Alexander MS, King PH. Smad8 Is Increased in Duchenne Muscular Dystrophy and Suppresses miR-1, miR-133a, and miR-133b. Int J Mol Sci 2022; 23:7515. [PMID: 35886863 PMCID: PMC9323105 DOI: 10.3390/ijms23147515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 12/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease characterized by skeletal muscle instability, progressive muscle wasting, and fibrosis. A major driver of DMD pathology stems from aberrant upregulation of transforming growth factor β (TGFβ) signaling. In this report, we investigated the major transducers of TGFβ signaling, i.e., receptor Smads (R-Smads), in DMD patient skeletal muscle and observed a 48-fold increase in Smad8 mRNA. Smad1, Smad2, Smad3, and Smad5 mRNA were only minimally increased. A similar pattern was observed in the muscle from the mdx5cv mouse. Western blot analysis showed upregulation of phosphorylated Smad1, Smad5, and Smad8 compared to total Smad indicating activation of this pathway. In parallel, we observed a profound diminishment of muscle-enriched microRNAs (myomiRs): miR-1, miR-133a, and miR-133b. The pattern of Smad8 induction and myomiR suppression was recapitulated in C2C12 muscle cells after stimulation with bone morphogenetic protein 4 (BMP4), a signaling factor that we found upregulated in DMD muscle. Silencing Smad8 in C2C12 myoblasts derepressed myomiRs and promoted myoblast differentiation; there was also a concomitant upregulation of myogenic regulatory factors (myogenin and myocyte enhancer factor 2D) and suppression of a pro-inflammatory cytokine (interleukin-6). Our data suggest that Smad8 is a negative regulator of miR-1, miR-133a, and miR-133b in muscle cells and that the BMP4-Smad8 axis is a driver of dystrophic pathology in DMD.
Collapse
Affiliation(s)
- Michael A. Lopez
- Children’s of Alabama, Birmingham, AL 35233, USA; (M.C.); (S.G.); (M.S.A.)
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
- Department of Neurology, University of Alabama at Birmingham (UAB), Civitan 545C, 1530 3rd Avenue South, Birmingham, AL 35294, USA; (Y.S.); (L.A.); (M.M.B.)
- UAB Center for Exercise Medicine (UCEM), University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
| | - Ying Si
- Department of Neurology, University of Alabama at Birmingham (UAB), Civitan 545C, 1530 3rd Avenue South, Birmingham, AL 35294, USA; (Y.S.); (L.A.); (M.M.B.)
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| | - Xianzhen Hu
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
| | - Valentyna Williams
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
| | - Fuad Qushair
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
| | - Jackson Carlyle
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
| | - Lyndsy Alesce
- Department of Neurology, University of Alabama at Birmingham (UAB), Civitan 545C, 1530 3rd Avenue South, Birmingham, AL 35294, USA; (Y.S.); (L.A.); (M.M.B.)
| | - Michael Conklin
- Children’s of Alabama, Birmingham, AL 35233, USA; (M.C.); (S.G.); (M.S.A.)
- Department of Orthopedic Surgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
| | - Shawn Gilbert
- Children’s of Alabama, Birmingham, AL 35233, USA; (M.C.); (S.G.); (M.S.A.)
- Department of Orthopedic Surgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
| | - Marcas M. Bamman
- Department of Neurology, University of Alabama at Birmingham (UAB), Civitan 545C, 1530 3rd Avenue South, Birmingham, AL 35294, USA; (Y.S.); (L.A.); (M.M.B.)
- UAB Center for Exercise Medicine (UCEM), University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
- Department of Cell, Development and Integrative Biology, Birmingham, AL 35233, USA
| | - Matthew S. Alexander
- Children’s of Alabama, Birmingham, AL 35233, USA; (M.C.); (S.G.); (M.S.A.)
- Department of Pediatrics, University of Alabama at Birmingham (UAB), CHB314, 1600 7th Avenue South, Birmingham, AL 35233, USA; (X.H.); (V.W.); (F.Q.); (J.C.)
- UAB Center for Exercise Medicine (UCEM), University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
- UAB Civitan International Research Center (CIRC), Birmingham, AL 35233, USA
- Department of Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
| | - Peter H. King
- Department of Neurology, University of Alabama at Birmingham (UAB), Civitan 545C, 1530 3rd Avenue South, Birmingham, AL 35294, USA; (Y.S.); (L.A.); (M.M.B.)
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
21
|
Rodrigues BM, Mathias LS, Deprá IDC, Cury SS, de Oliveira M, Olimpio RMC, De Sibio MT, Gonçalves BM, Nogueira CR. Effects of Triiodothyronine on Human Osteoblast-Like Cells: Novel Insights From a Global Transcriptome Analysis. Front Cell Dev Biol 2022; 10:886136. [PMID: 35784485 PMCID: PMC9248766 DOI: 10.3389/fcell.2022.886136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Thyroid hormones play a significant role in bone development and maintenance, with triiodothyronine (T3) particularly being an important modulator of osteoblast differentiation, proliferation, and maintenance. However, details of the biological processes (BPs) and molecular pathways affected by T3 in osteoblasts remain unclear.Methods: To address this issue, primary cultures of human adipose-derived mesenchymal stem cells were subjected to our previously established osteoinduction protocol, and the resultant osteoblast-like cells were treated with 1 nm or 10 nm T3 for 72 h. RNA sequencing (RNA-Seq) was performed using the Illumina platform, and differentially expressed genes (DEGs) were identified from the raw data using Kallisto and DESeq2. Enrichment analysis of DEGs was performed against the Gene Ontology Consortium database for BP terms using the R package clusterProfiler and protein network analysis by STRING.Results: Approximately 16,300 genes were analyzed by RNA-Seq, with 343 DEGs regulated in the 1 nm T3 group and 467 upregulated in the 10 nm T3 group. Several independent BP terms related to bone metabolism were significantly enriched, with a number of genes shared among them (FGFR2, WNT5A, WNT3, ROR2, VEGFA, FBLN1, S1PR1, PRKCZ, TGFB3, and OSR1 for 1nM T3; and FZD1, SMAD6, NOG, NEO1, and ENG for 10 nm T3). An osteoblast-related search in the literature regarding this set of genes suggests that both T3 doses are unfavorable for osteoblast development, mainly hindering BMP and canonical and non-canonical WNT signaling.Conclusions: Therefore, this study provides new directions toward the elucidation of the mechanisms of T3 action on osteoblast metabolism, with potential future implications for the treatment of endocrine-related bone pathologies.
Collapse
Affiliation(s)
- Bruna Moretto Rodrigues
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Lucas Solla Mathias
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Igor de Carvalho Deprá
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Sarah Santiloni Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Miriane de Oliveira
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Maria Teresa De Sibio
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Bianca Mariani Gonçalves
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Célia Regina Nogueira
- Department of Internal Medicine, Medical School Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
- *Correspondence: Célia Regina Nogueira,
| |
Collapse
|
22
|
Huang CC, Hung CH, Lee YJ, Tseng TH, Lee YJ, Kao SH, Wang CJ. Camphorataimide B suppresses the metastasis of human colorectal cancer cell by inhibiting Smad/FAK/Akt axis and promoting degradation of Snail/BMP4 complex. J Food Drug Anal 2022; 30:271-282. [PMID: 39666299 PMCID: PMC9635903 DOI: 10.38212/2224-6614.3405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2024] Open
Abstract
Camphorataimide B (CamB) has anticancer activities against several tumors. Here, we aimed to investigate the mechanism(s) by which CamB inhibits metastasis of colorectal cancer (CRC) cells. Low-dose CamB did not affect the cell viability and cell cycle progression of CRC cells, but significantly inhibited the metastatic potentials of CRC cells. Mechanically, CamB reduced the protein and mRNA expression of BMP4, and inhibited Smad and FAK/Src/Akt signaling. CamB also decreased Snail levels by promoting its degradation via proteasome and thereafter reduced Snail-mediated BMP4 transcription. Moreover, CamB considerably inhibited the in vivo metastasis of DLD-1 cells in xenograft mice.
Collapse
Affiliation(s)
- Chi-Chou Huang
- Department of Colorectal Surgery, Chung Shan Medical University Hospital, Taichung,
Taiwan
- School of Medicine, Chung Shan Medical University, Taichung,
Taiwan
| | - Chia-Hung Hung
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung,
Taiwan
| | - Yi-Ju Lee
- Department of Pathology, Chung Shan Medical University Hospital, Taichung,
Taiwan
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung,
Taiwan
| | - Tsui-Hwa Tseng
- Department of Medical Chemistry, Chung Shan Medical University, Taichung,
Taiwan
| | - Yean-Jang Lee
- Department of Chemistry, National Changhua University of Education, Changhua,
Taiwan
| | - Shao-Hsuan Kao
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung,
Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung,
Taiwan
| | - Chau-Jong Wang
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung,
Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung,
Taiwan
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung,
Taiwan
| |
Collapse
|
23
|
Ehata S, Miyazono K. Bone Morphogenetic Protein Signaling in Cancer; Some Topics in the Recent 10 Years. Front Cell Dev Biol 2022; 10:883523. [PMID: 35693928 PMCID: PMC9174896 DOI: 10.3389/fcell.2022.883523] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/09/2022] [Indexed: 12/19/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-β (TGF-β) family, are multifunctional cytokines. BMPs have a broad range of functions, and abnormalities in BMP signaling pathways are involved in cancer progression. BMPs activate the proliferation of certain cancer cells. Malignant phenotypes of cancer cells, such as increased motility, invasiveness, and stemness, are enhanced by BMPs. Simultaneously, BMPs act on various cellular components and regulate angiogenesis in the tumor microenvironment. Thus, BMPs function as pro-tumorigenic factors in various types of cancer. However, similar to TGF-β, which shows both positive and negative effects on tumorigenesis, BMPs also act as tumor suppressors in other types of cancers. In this article, we review important findings published in the recent decade and summarize the pro-oncogenic functions of BMPs and their underlying mechanisms. The current status of BMP-targeted therapies for cancers is also discussed.
Collapse
Affiliation(s)
- Shogo Ehata
- Department of Pathology, School of Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Shogo Ehata,
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Ukan Ü, Delgado Lagos F, Kempf S, Günther S, Siragusa M, Fisslthaler B, Fleming I. Effect of Thrombin on the Metabolism and Function of Murine Macrophages. Cells 2022; 11:cells11101718. [PMID: 35626753 PMCID: PMC9139186 DOI: 10.3390/cells11101718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophages are plastic and heterogeneous immune cells that adapt pro- or anti-inflammatory phenotypes upon exposure to different stimuli. Even though there has been evidence supporting a crosstalk between coagulation and innate immunity, the way in which protein components of the hemostasis pathway influence macrophages remains unclear. We investigated the effect of thrombin on macrophage polarization. On the basis of gene expression and cytokine secretion, our results suggest that polarization with thrombin induces an anti-inflammatory, M2-like phenotype. In functional studies, thrombin polarization promoted oxLDL phagocytosis by macrophages, and conditioned medium from the same cells increased endothelial cell proliferation. There were, however, clear differences between the classical M2a polarization and the effects of thrombin on gene expression. Finally, the deletion and inactivation of secreted modular Ca2+-binding protein 1 (SMOC1) attenuated phagocytosis by thrombin-stimulated macrophages, a phenomenon revered by the addition of recombinant SMOC1. Manipulation of SMOC1 levels also had a pronounced impact on the expression of TGF-β-signaling-related genes. Taken together, our results show that thrombin induces an anti-inflammatory macrophage phenotype with similarities as well as differences to the classical alternatively activated M2 polarization states, highlighting the importance of tissue levels of SMOC1 in modifying thrombin-induced macrophage polarization.
Collapse
Affiliation(s)
- Ürün Ukan
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Fredy Delgado Lagos
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Sebastian Kempf
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Beate Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, 60596 Frankfurt am Main, Germany
- CardioPulmonary Institute, Goethe University, 60596 Frankfurt am Main, Germany
- Correspondence:
| |
Collapse
|
25
|
Yang S, Ning G, Hou Y, Cao Y, Xu J, Wu J, Zhang T, Wang Q. Myoneurin regulates BMP signaling by competing with Ppm1a for Smad binding. iScience 2022; 25:104495. [PMID: 35712083 PMCID: PMC9194458 DOI: 10.1016/j.isci.2022.104495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
A delicate balance of BMP activity is critical for tissue formation and organogenesis. However, the mechanical molecular details in ensuring the proper duration and intensity of BMP signaling have yet to be fully elucidated. Here, we identified a zebrafish mutant with a disrupted gene encoding for the BTB/POZ and zinc finger protein myoneurin (Mynn). mynn−/− mutants exhibited severe loss of pharyngeal cartilage elements, owing to poor proliferation, blocked differentiation, and low viability of cranial neural crest cells. Depletion of mynn in both zebrafish embryos and mammalian cells led to a reduction of the BMP signal activity. Mechanistically, Mynn interacts with Smad proteins in the nucleus, thereby disrupting the association between Smad protein and the phosphatase Ppm1a. Ultimately, this interaction prevents Smad dephosphorylation. More broadly, our findings may provide a new strategy to balance BMP signal activity via competitive binding of Mynn and Ppm1a to Smad proteins during pharyngeal cartilage formation. mynn gene is essential for pharyngeal cartilage development mynn is required for the proliferation, differentiation, and survival of the CNCCs Mynn has an evolutionarily conserved function in supporting BMP signal Mynn maintains BMP signal activity by competing with Ppm1a for Smad binding
Collapse
|
26
|
Garg B, Tomar N, Biswas A, Mehta N, Malhotra R. Understanding Musculoskeletal Disorders Through Next-Generation Sequencing. JBJS Rev 2022; 10:01874474-202204000-00001. [PMID: 35383688 DOI: 10.2106/jbjs.rvw.21.00165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
» An insight into musculoskeletal disorders through advancements in next-generation sequencing (NGS) promises to maximize benefits and improve outcomes through improved genetic diagnosis. » The primary use of whole exome sequencing (WES) for musculoskeletal disorders is to identify functionally relevant variants. » The current evidence has shown the superiority of NGS over conventional genotyping for identifying novel and rare genetic variants in patients with musculoskeletal disorders, due to its high throughput and low cost. » Genes identified in patients with scoliosis, osteoporosis, osteoarthritis, and osteogenesis imperfecta using NGS technologies are listed for further reference.
Collapse
Affiliation(s)
- Bhavuk Garg
- Department of Orthopaedics, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | |
Collapse
|
27
|
Kaminker JD, Timoshenko AV. Expression, Regulation, and Functions of the Galectin-16 Gene in Human Cells and Tissues. Biomolecules 2021; 11:1909. [PMID: 34944551 PMCID: PMC8699332 DOI: 10.3390/biom11121909] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022] Open
Abstract
Galectins comprise a family of soluble β-galactoside-binding proteins, which regulate a variety of key biological processes including cell growth, differentiation, survival, and death. This paper aims to address the current knowledge on the unique properties, regulation, and expression of the galectin-16 gene (LGALS16) in human cells and tissues. To date, there are limited studies on this galectin, with most focusing on its tissue specificity to the placenta. Here, we report the expression and 8-Br-cAMP-induced upregulation of LGALS16 in two placental cell lines (BeWo and JEG-3) in the context of trophoblastic differentiation. In addition, we provide the results of a bioinformatics search for LGALS16 using datasets available at GEO, Human Protein Atlas, and prediction tools for relevant transcription factors and miRNAs. Our findings indicate that LGALS16 is detected by microarrays in diverse human cells/tissues and alters expression in association with cancer, diabetes, and brain diseases. Molecular mechanisms of the transcriptional and post-transcriptional regulation of LGALS16 are also discussed based on the available bioinformatics resources.
Collapse
|
28
|
Jin Q, Jiang X, Du X, Hu W, Bai S, Wang X, Xu B, Zhao W. Integrated Transcriptome and Multiple Activated Pathways in Endometrial Cancer. Front Genet 2021; 12:680331. [PMID: 34925436 PMCID: PMC8678463 DOI: 10.3389/fgene.2021.680331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 10/07/2021] [Indexed: 12/05/2022] Open
Abstract
Because the incidence of endometrial cancer is notably increasing worldwide, it has become the leading gynecologic cancer in the United States. Standard treatment results in the loss of reproductive function in women of childbearing age. Furthermore, advanced cancer stages are associated with poor overall survival. The aim of this study was to explore the abnormal expression profile of genes during the development of endometrial cancer, which is essential to provide a better understanding of the mechanisms involved. Five pairs of endometrial cancer tissues and normal endometrial tissues were subjected to next-generation transcriptome sequencing technology. Quantitative real-time PCR (RT-qPCR) was performed to validate the expression profile of key differentially expressed genes (2.0-fold change, adj. p < 0.05) (DEGs) identified in the RNA-seq result. GO and KEGG pathways were used for bioinformatic analyses. The transcriptomic sequencing results showed 1153 DEGs, including 673 upregulated and 480 downregulated genes, in the EC specimens. Decreased expression of ID1, IGF1, GDF7, SMAD9, TGF-beta and WNT4, as well as GDF5, INHBA and ERBB4 overexpression, were confirmed in EC using RT-qPCR. Additionally, EC tissue exhibited marked enrichment in genes promoting cellular adhesion, proliferation, migration and plasma membrane. KEGG analysis revealed changes in various pathways, such as the TGF-beta, PI3K-Akt, Wnt, and estrogen pathways. Our data describe the molecular events involved in the pathogenesis of EC, which may be potential diagnostic markers and targets of therapeutic interventions.
Collapse
Affiliation(s)
- Qi Jin
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiaohua Jiang
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xin Du
- Reproductive Medicine Center, 901th Hospital of PLA Joint Logistic Support Force, Hefei, China
| | - Weiping Hu
- Division of Life Sciences and Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Shun Bai
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xian Wang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Xu
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- *Correspondence: Bo Xu, ; Weidong Zhao,
| | - Weidong Zhao
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- *Correspondence: Bo Xu, ; Weidong Zhao,
| |
Collapse
|
29
|
Blanc V, Riordan JD, Soleymanjahi S, Nadeau JH, Nalbantoglu ILK, Xie Y, Molitor EA, Madison BB, Brunt EM, Mills JC, Rubin DC, Ng IO, Ha Y, Roberts LR, Davidson NO. Apobec1 complementation factor overexpression promotes hepatic steatosis, fibrosis, and hepatocellular cancer. J Clin Invest 2021; 131:138699. [PMID: 33445170 DOI: 10.1172/jci138699] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
The RNA-binding protein Apobec1 complementation factor (A1CF) regulates posttranscriptional ApoB mRNA editing, but the range of RNA targets and the long-term effect of altered A1CF expression on liver function are unknown. Here we studied hepatocyte-specific A1cf-transgenic (A1cf+/Tg), A1cf+/Tg Apobec1-/-, and A1cf-/- mice fed chow or high-fat/high-fructose diets using RNA-Seq, RNA CLIP-Seq, and tissue microarrays from human hepatocellular cancer (HCC). A1cf+/Tg mice exhibited increased hepatic proliferation and steatosis, with increased lipogenic gene expression (Mogat1, Mogat2, Cidea, Cd36) associated with shifts in polysomal RNA distribution. Aged A1cf+/Tg mice developed spontaneous fibrosis, dysplasia, and HCC, and this development was accelerated on a high-fat/high-fructose diet and was independent of Apobec1. RNA-Seq revealed increased expression of mRNAs involved in oxidative stress (Gstm3, Gpx3, Cbr3), inflammatory response (Il19, Cxcl14, Tnfα, Ly6c), extracellular matrix organization (Mmp2, Col1a1, Col4a1), and proliferation (Kif20a, Mcm2, Mcm4, Mcm6), and a subset of mRNAs (including Sox4, Sox9, Cdh1) were identified in RNA CLIP-Seq. Increased A1CF expression in human HCC correlated with advanced fibrosis and with reduced survival in a subset with nonalcoholic fatty liver disease. In conclusion, we show that hepatic A1CF overexpression selectively alters polysomal distribution and mRNA expression, promoting lipogenic, proliferative, and inflammatory pathways leading to HCC.
Collapse
Affiliation(s)
- Valerie Blanc
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jesse D Riordan
- Pacific Northwest Research Institute, Seattle, Washington, USA
| | - Saeed Soleymanjahi
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph H Nadeau
- Pacific Northwest Research Institute, Seattle, Washington, USA
| | - ILKe Nalbantoglu
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yan Xie
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth A Molitor
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Blair B Madison
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth M Brunt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jason C Mills
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irene O Ng
- Department of Pathology and State Key Laboratory of Liver Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Yeonjung Ha
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Lewis R Roberts
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Watanabe N, Nakano M, Mitsuishi Y, Hara N, Mano T, Iwata A, Murayama S, Suzuki T, Ikeuchi T, Nishimura M. Transcriptional downregulation of FAM3C/ILEI in the Alzheimer's brain. Hum Mol Genet 2021; 31:122-132. [PMID: 34378027 DOI: 10.1093/hmg/ddab226] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 11/14/2022] Open
Abstract
Amyloid-β (Aβ) accumulation in the brain triggers the pathogenic cascade for Alzheimer's disease (AD) development. The secretory protein FAM3C (also named ILEI) is a candidate for an endogenous suppressor of Aβ production. In this study, we found that FAM3C expression was transcriptionally downregulated in the AD brain. To determine the transcriptional mechanism of the human FAM3C gene, we delineated the minimal 5'-flanking sequence required for basal promoter activity. From a database search for DNA-binding motifs, expression analysis using cultured cells, and promoter DNA-binding assays, we identified SP1 and EBF1 as candidate basal transcription factors for FAM3C, and found that SMAD1 was a putative inducible transcription factor and KLF6 was a transcription repressor for FAM3C. Genomic deletion of the basal promoter sequence from HEK293 and Neuro-2a cells markedly reduced endogenous expression of FAM3C and abrogated SP1- or EBF1-mediated induction of FAM3C. Nuclear protein extracts from AD brains contained lower levels of SP1 and EBF1 than did those from control brains, although the relative mRNA levels of these factors did not differ significantly between the groups. Additionally, the ability of nuclear SP1 and EBF1 in AD brains to bind with the basal promoter sequence-containing DNA probe was reduced compared with the binding ability of these factors in control brains. Thus, the transcriptional downregulation of FAM3C in the AD brain is attributable to the reduced nuclear levels and genomic DNA binding of SP1 and EBF1. An expressional decline in FAM3C may be a risk factor for Aβ accumulation and eventually AD development.
Collapse
Affiliation(s)
- Naoki Watanabe
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Masaki Nakano
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Yachiyo Mitsuishi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Tatsuo Mano
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Atsushi Iwata
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Hokkaido 060-0812, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| |
Collapse
|
31
|
Li YD, Liu X, Li ZW, Wang WJ, Li YM, Cao ZP, Luan P, Xiao F, Gao HH, Guo HS, Wang N, Li H, Wang SZ. A combination of genome-wide association study and selection signature analysis dissects the genetic architecture underlying bone traits in chickens. Animal 2021; 15:100322. [PMID: 34311193 DOI: 10.1016/j.animal.2021.100322] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/01/2023] Open
Abstract
The bones of chicken play an important role in supporting and protecting the body. The growth and development of bones have a substantial influence on the health and production performance in chickens. However, genetic architecture underlying chicken bone traits is not well understood. The objectives of this study are to dissect the genetic basis of bone traits in chickens and to identify valuable genes and genetic markers for chicken breeding. We performed a combination of genome-wide association study (GWAS) and selection signature analysis (fixation index values and nucleotide diversity ratios) in an F2 crossbred experimental population with different genetic backgrounds (broiler × layer) to identify candidate genes and significant variants related to femur, shank, keel length, chest width, metatarsal claw weight, metatarsal length, and metatarsal circumference. A total of 545 individuals were genotyped based on the whole genome re-sequencing method (26 F0 individuals were re-sequenced at 10 × coverage; 519 F2 individuals were re-sequenced at 3 × coverage). A total of 2 028 112 single-nucleotide polymorphisms (SNPs) remained to carry out analysis after quality control and imputation. The integration of GWAS and selection signature analysis indicated that all significant SNPs responsible for bone traits were mainly localized on chicken chromosomes 1, 4, and 27. Finally, we identified 21 positional candidate genes that might regulate chicken bone growth and development, including LRCH1, RB1, FNDC3A, MLNR, CAB39L, FOXO1, LHFP, TRPC4, POSTN, SMAD9, RBPJ, PPARGC1A, SLIT2, NCAPG, NKX3-2, CPZ, SPOP, NGFR, SOST, ZNF652, and HOXB3. Additionally, an array of uncharacterized genes was identified. The findings provide an in-depth understanding of the genetic architecture of chicken bone traits and offer a molecular basis for applying genomics in practical chicken breeding.
Collapse
Affiliation(s)
- Y D Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - X Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Z W Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - W J Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Y M Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Z P Cao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - P Luan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - F Xiao
- Fujian Sunnzer Biotechnology Development Co., Ltd, Guangze, Fujian Province 354100, PR China
| | - H H Gao
- Fujian Sunnzer Biotechnology Development Co., Ltd, Guangze, Fujian Province 354100, PR China
| | - H S Guo
- Fujian Sunnzer Biotechnology Development Co., Ltd, Guangze, Fujian Province 354100, PR China
| | - N Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - H Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - S Z Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
32
|
Aashaq S, Batool A, Mir SA, Beigh MA, Andrabi KI, Shah ZA. TGF-β signaling: A recap of SMAD-independent and SMAD-dependent pathways. J Cell Physiol 2021; 237:59-85. [PMID: 34286853 DOI: 10.1002/jcp.30529] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
Transforming growth factor-β (TGF-β) is a proinflammatory cytokine known to control a diverse array of pathological and physiological conditions during normal development and tumorigenesis. TGF-β-mediated physiological effects are heterogeneous and vary among different types of cells and environmental conditions. TGF-β serves as an antiproliferative agent and inhibits tumor development during primary stages of tumor progression; however, during the later stages, it encourages tumor development and mediates metastatic progression and chemoresistance. The fundamental elements of TGF-β signaling have been divulged more than a decade ago; however, the process by which the signals are relayed from cell surface to nucleus is very complex with additional layers added in tumor cell niches. Although the intricate understanding of TGF-β-mediated signaling pathways and their regulation are still evolving, we tried to make an attempt to summarize the TGF-β-mediated SMAD-dependent andSMAD-independent pathways. This manuscript emphasizes the functions of TGF-β as a metastatic promoter and tumor suppressor during the later and initial phases of tumor progression respectively.
Collapse
Affiliation(s)
- Sabreena Aashaq
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| | - Asiya Batool
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine, Srinagar, JK, India
| | | | | | | | - Zaffar Amin Shah
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| |
Collapse
|
33
|
Zhang HW, Guo Y, Sun LX, Ni FB, Xu K. Prognostic value of small mother against decapentaplegic expression in human gastric cancer. Bioengineered 2021; 12:2534-2549. [PMID: 34138687 PMCID: PMC8806811 DOI: 10.1080/21655979.2021.1935192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gastric cancer is the fifth most common malignancy in the world with alow 5-year survival rate. To date, no study has investigated the prognostic role of the small mother against decapentaplegic (SMAD) in gastric cancer. The association of SMADs with overall survival (OS) of gastric cancer was analyzed on the online Kaplan-Meier (KM) plotter database. Clinical data such as stage, differentiation, gender, treatment, and Her2 mutation status of gastric cancer patients were analyzed. The (E)-SIS3 was used to inhibit SMAD3 expression in gastric cancer cells, and the effects of SMAD3 on gastric cancer cells were analyzed via real-time cellular analysis (RTCA), flow cytometry, colony formation, and immunofluorescence assay. The results showed that the high expression of three members of SMADs (SMAD1, SMAD2, SMAD4) was correlated with afavorable OS of gastric cancer patients. Meanwhile, SMAD3 expression level indicated highly differentiated cancer. We also observed that surgical treatment was associated with high expression level of SMAD1 and SMAD2. Besides, the effect of Her2 on gastric cancer was not noticeable. Moreover, (E)-SIS3 pharmacological assay revealed that inhibition of expression of SMAD3 suppressed the proliferation and migration ability of gastric cancer cells via inducing apoptosis. Collectively, these results demonstrate that the high expression level of three members of SMADs (SMAD1, SMAD2, and SMAD4) is significantly correlated with favorable OS of gastric cancer patients, which is opposite to SMAD3. Thus, SMADs regulate the differentiation of cancer and can be used to guide treatment decisions.
Collapse
Affiliation(s)
- He-Wei Zhang
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Ying Guo
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Lin-Xiao Sun
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fu-Biao Ni
- Department of General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Ke Xu
- Endocrinology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| |
Collapse
|
34
|
Yang D, Yang X, Dai F, Wang Y, Yang Y, Hu M, Cheng Y. The Role of Bone Morphogenetic Protein 4 in Ovarian Function and Diseases. Reprod Sci 2021; 28:3316-3330. [PMID: 33966186 DOI: 10.1007/s43032-021-00600-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β (TGF-β) superfamily. BMP4 is a secreted protein that was originally identified due to its role in bone and cartilage development. Over the past decades, extensive literature has indicated that BMP4 and its receptors are widely expressed in the ovary. Dysregulation of BMP4 expression may play a vital role in follicular development, polycystic ovary syndrome (PCOS), and ovarian cancer. In this review, we summarized the expression pattern of BMP4 in the ovary, focused on the role of BMP4 in follicular development and steroidogenesis, and discussed the role of BMP4 in ovarian diseases such as polycystic ovary syndrome and ovarian cancer. Some studies have shown that the expression of BMP4 in the ovary is spatiotemporal and species specific, but the effects of BMP4 seem to be similar in follicular development of different species. In addition, BMP4 is involved in the development of hyperandrogenemia in PCOS and drug resistance in ovarian cancer, but further research is still needed to clarify the specific mechanisms.
Collapse
Affiliation(s)
- Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yang
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan, 430072, China.
| | - Min Hu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
35
|
Hodgson J, Ruiz-Llorente L, McDonald J, Quarrell O, Ugonna K, Bentham J, Mason R, Martin J, Moore D, Bergstrom K, Bayrak-Toydemir P, Wooderchak-Donahue W, Morrell NW, Condliffe R, Bernabeu C, Upton PD. Homozygous GDF2 nonsense mutations result in a loss of circulating BMP9 and BMP10 and are associated with either PAH or an "HHT-like" syndrome in children. Mol Genet Genomic Med 2021; 9:e1685. [PMID: 33834622 PMCID: PMC8683697 DOI: 10.1002/mgg3.1685] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 12/28/2022] Open
Abstract
Background Disrupted endothelial BMP9/10 signaling may contribute to the pathophysiology of both hereditary hemorrhagic telangiectasia (HHT) and pulmonary arterial hypertension (PAH), yet loss of circulating BMP9 has not been confirmed in individuals with ultra‐rare homozygous GDF2 (BMP9 gene) nonsense mutations. We studied two pediatric patients homozygous for GDF2 (BMP9 gene) nonsense mutations: one with PAH (c.[76C>T];[76C>T] or p.[Gln26Ter];[Gln26Ter] and a new individual with pulmonary arteriovenous malformations (PAVMs; c.[835G>T];[835G>T] or p.[Glu279Ter];[Glu279Ter]); both with facial telangiectases. Methods Plasma samples were assayed for BMP9 and BMP10 by ELISA. In parallel, serum BMP activity was assayed using an endothelial BRE‐luciferase reporter cell line (HMEC1‐BRE). Proteins were expressed for assessment of secretion and processing. Results Plasma levels of both BMP9 and BMP10 were undetectable in the two homozygous index cases and this corresponded to low serum‐derived endothelial BMP activity in the patients. Measured BMP9 and BMP10 levels were reduced in the asymptomatic heterozygous p.[Glu279Ter] parents, but serum activity was normal. Although expression studies suggested alternate translation can be initiated at Met57 in the p.[Gln26Ter] mutant, this does not result in secretion of functional BMP9. Conclusion Collectively, these data show that homozygous GDF2 mutations, leading to a loss of circulating BMP9 and BMP10, can cause either pediatric PAH and/or “HHT‐like” telangiectases and PAVMs. Although patients reported to date have manifestations that overlap with those of HHT, none meet the Curaçao criteria for HHT and seem distinct from HHT in terms of the location and appearance of telangiectases, and a tendency for tiny, diffuse PAVMs.
Collapse
Affiliation(s)
- Joshua Hodgson
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lidia Ruiz-Llorente
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Department of Systems Biology, School of Medicine and Health Sciences, University of Alcalá, Madrid, Spain
| | - Jamie McDonald
- HHT Center, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Oliver Quarrell
- Department of Clinical Genetics, Sheffield Children's Hospital, Sheffield, UK
| | - Kelechi Ugonna
- Department of Respiratory Medicine, Sheffield Children's Hospital, Sheffield, UK
| | - James Bentham
- Department of Paediatric Congenital Heart Disease, Leeds Children's Hospital, Leeds, UK
| | - Rebecca Mason
- Department of Clinical Genetics, Sheffield Children's Hospital, Sheffield, UK
| | - Jennifer Martin
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - David Moore
- NHS Lothian Molecular Genetics Service, Western General Hospital, Edinburgh, UK
| | - Katie Bergstrom
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | | | | | | | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Paul D Upton
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Refolding, purification, and characterization of constitutive-active human-Smad8 produced as inclusion bodies in ClearColi® BL21 (DE3). Protein Expr Purif 2021; 184:105878. [PMID: 33812004 DOI: 10.1016/j.pep.2021.105878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 11/21/2022]
Abstract
Smad8 is a transcriptional regulator that participates in the intracellular signaling pathway of the transforming growth factor-β (TGF-β) family. Full-length Smad8 is an inactive protein in the absence of ligand stimulation. The expression of a truncated version of the protein lacking the MH1 domain (cSmad8) revealed constitutive activity in genetically engineered mesenchymal stem cells and, in combination with BMP-2, exhibited a tendon cell-inducing potential. To further explore function and applicability of Smad8 in regenerative medicine recombinant production is required. Herein, we further engineered cSmad8 to include the transactivation signal (TAT) of the human immunodeficiency virus (HIV) to allow internalization into cells. TAT-hcSmad8 was produced in endotoxin-free ClearColi® BL21 (DE3), refolded from inclusion bodies (IBs) and purified by Heparin chromatography. Analysis of TAT-hcSmad8 by thermal shift assay revealed the formation of a hydrophobic core. The presence of mixed α-helixes and β-sheets, in line with theoretical models, was proven by circular dichroism. TAT-hcSmad8 was successfully internalized by C3H10T1/2 cells, where it was mainly found in the cytoplasm and partially in the nucleus. Finally, it was shown that TAT-hcSmad8 exhibited biological activity in C3H10T1/2 cells after co-stimulation with BMP-2.
Collapse
|
37
|
Racine C, Genêt C, Bourgneuf C, Dupont C, Plisson-Petit F, Sarry J, Hennequet-Antier C, Vigouroux C, Mathieu d'Argent E, Pierre A, Monniaux D, Fabre S, di Clemente N. New Anti-Müllerian Hormone Target Genes Involved in Granulosa Cell Survival in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2021; 106:e1271-e1289. [PMID: 33247926 DOI: 10.1210/clinem/dgaa879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE A protective effect of anti-Müllerian hormone (AMH) on follicle atresia was recently demonstrated using long-term treatments, but this effect has never been supported by mechanistic studies. This work aimed to gain an insight into the mechanism of action of AMH on follicle atresia and on how this could account for the increased follicle pool observed in women with polycystic ovary syndrome (PCOS). METHODS In vivo and in vitro experiments were performed to study the effects of AMH on follicle atresia and on the proliferation and apoptosis of granulosa cells (GCs). RNA-sequencing was carried out to identify new AMH target genes in GCs. The expression of some of these genes in GCs from control and PCOS women was compared using microfluidic real time quantitative RT-PCR. RESULTS A short-term AMH treatment prevented follicle atresia in prepubertal mice. Consistent with this result, AMH inhibited apoptosis and promoted proliferation of different models of GCs. Moreover, integrative biology analyses of 965 AMH target genes identified in 1 of these GC models, confirmed that AMH had initiated a gene expression program favoring cell survival and proliferation. Finally, on 43 genes selected among the most up- and down-regulated AMH targets, 8 were up-regulated in GCs isolated from PCOS women, of which 5 are involved in cell survival. MAIN CONCLUSIONS Our results provide for the first time cellular and molecular evidence that AMH protects follicles from atresia by controlling GC survival and suggest that AMH could participate in the increased follicle pool of PCOS patients.
Collapse
Affiliation(s)
- Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Carine Genêt
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Camille Bourgneuf
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Charlotte Dupont
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | | | - Julien Sarry
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Christelle Hennequet-Antier
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Corinne Vigouroux
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Emmanuelle Mathieu d'Argent
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Danielle Monniaux
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Stéphane Fabre
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
38
|
Pharmacological Manipulation of Early Zebrafish Skeletal Development Shows an Important Role for Smad9 in Control of Skeletal Progenitor Populations. Biomolecules 2021; 11:biom11020277. [PMID: 33668680 PMCID: PMC7918065 DOI: 10.3390/biom11020277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/16/2022] Open
Abstract
Osteoporosis and other conditions associated with low bone density or quality are highly prevalent, are increasing as the population ages and with increased glucocorticoid use to treat conditions with elevated inflammation. There is an unmet need for therapeutics which can target skeletal precursors to induce osteoblast differentiation and osteogenesis. Genes associated with high bone mass represent interesting targets for manipulation, as they could offer ways to increase bone density. A damaging mutation in SMAD9 has recently been associated with high bone mass. Here we show that Smad9 labels groups of osteochondral precursor cells, which are not labelled by the other Regulatory Smads: Smad1 or Smad5. We show that Smad9+ cells are proliferative, and that the Smad9+ pocket expands following osteoblast ablation which induced osteoblast regeneration. We further show that treatment with retinoic acid, prednisolone, and dorsomorphin all alter Smad9 expression, consistent with the effects of these drugs on the skeletal system. Taken together these results demonstrate that Smad9+ cells represent an undifferentiated osteochondral precursor population, which can be manipulated by commonly used skeletal drugs. We conclude that Smad9 represents a target for future osteoanabolic therapies.
Collapse
|
39
|
Nagel S, Pommerenke C, Meyer C, MacLeod RAF, Drexler HG. Establishment of the TALE-code reveals aberrantly activated homeobox gene PBX1 in Hodgkin lymphoma. PLoS One 2021; 16:e0246603. [PMID: 33539429 PMCID: PMC7861379 DOI: 10.1371/journal.pone.0246603] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
Homeobox genes encode transcription factors which regulate basic processes in development and cell differentiation and are grouped into classes and subclasses according to sequence similarities. Here, we analyzed the activities of the 20 members strong TALE homeobox gene class in early hematopoiesis and in lymphopoiesis including developing and mature B-cells, T-cells, natural killer (NK)-cells and innate lymphoid cells (ILC). The resultant expression pattern comprised eleven genes and which we termed TALE-code enables discrimination of normal and aberrant activities of TALE homeobox genes in lymphoid malignancies. Subsequent expression analysis of TALE homeobox genes in public datasets of Hodgkin lymphoma (HL) patients revealed overexpression of IRX3, IRX4, MEIS1, MEIS3, PBX1, PBX4 and TGIF1. As paradigm we focused on PBX1 which was deregulated in about 17% HL patients. Normal PBX1 expression was restricted to hematopoietic stem cells and progenitors of T-cells and ILCs but absent in B-cells, reflecting its roles in stemness and early differentiation. HL cell line SUP-HD1 expressed enhanced PBX1 levels and served as an in vitro model to identify upstream regulators and downstream targets in this malignancy. Genomic studies of this cell line therein showed a gain of the PBX1 locus at 1q23 which may underlie its aberrant expression. Comparative expression profiling analyses of HL patients and cell lines followed by knockdown experiments revealed NFIB and TLX2 as target genes activated by PBX1. HOX proteins operate as cofactors of PBX1. Accordingly, our data showed that HOXB9 overexpressed in HL coactivated TLX2 but not NFIB while activating TNFRSF9 without PBX1. Further downstream analyses showed that TLX2 activated TBX15 which operated anti-apoptotically. Taken together, we discovered a lymphoid TALE-code and identified an aberrant network around deregulated TALE homeobox gene PBX1 which may disturb B-cell differentiation in HL by reactivation of progenitor-specific genes. These findings may provide the framework for future studies to exploit possible vulnerabilities of malignant cells in therapeutic scenarios.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Roderick A. F. MacLeod
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Hans G. Drexler
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ – German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
40
|
Gao L, Tian Q, Wu T, Shi S, Yin X, Liu L, Zheng L, Wang P, Tian Y, Xu S. Reduction of miR-744 delivered by NSCLC cell-derived extracellular vesicles upregulates SUV39H1 to promote NSCLC progression via activation of the Smad9/BMP9 axis. J Transl Med 2021; 19:37. [PMID: 33472665 PMCID: PMC7816389 DOI: 10.1186/s12967-020-02654-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is a common type of lung cancer. Extracellular vehicles (EVs) are nano-sized particles containing proteins, lipids, and miRNAs secreted by various cells, which play important roles in the development of lung cancer by regulating a wide range of signaling pathways. This study focused on elucidating a potential mechanism by which EVs promote the development of NSCLC. Methods Expression levels of miR-744, SUV39H1, Smad9, and BMP4 in clinical tissue samples of NSCLC patients and cell lines were quantified by RT-qPCR and/or western blot analysis. The interaction between SUV39H1 and miR-744 was identified by dual-luciferase reporter assay. miR-744, SUV39H1, and BMP4 expression levels were modulated in A549 and H460 cells, in order to evaluate their effects on cell proliferation, colony formation and cell cycle. A NSCLC xenograft mouse model was used to verify the in vitro findings. NSCLC cell-derived EVs and normal bronchial epithelial cell-derived EVs were isolated and their roles in NSCLC development were evaluated in vivo and in vitro. Results miR-744 expression was lower in cancer cell-derived derived EVs than in normal lung epithelial cell-derived EVs. Reduced miR-744 expression in EVs upregulated SUV39H1 in NSCLC cells and further increased BMP4 levels to promote NSCLC development. BMP4 was upregulated in NSCLC cells upon suppression of Smad9 mediated by SUV39H1. Reduced miR-744 expression transferred from cancer cell-derived EVs into NSCLC cells enhanced cancer development. Conclusion Overall, our findings unveiled a mechanism whereby miR-744 delivered by NSCLC-derived EVs upregulated SUV39H1, activating the Smad9/BMP9 axis and thus promoted cancer development.
Collapse
Affiliation(s)
- Liming Gao
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, P.R. China
| | - Qi Tian
- Department of Respiratory, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, P.R. China
| | - Tong Wu
- Hebei Medical University, Shijiazhuang, 050000, P.R. China
| | - Shanshan Shi
- Department of Respiratory, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, P.R. China
| | - Xiaobo Yin
- Department of Respiratory, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, P.R. China
| | - Lijie Liu
- Chengde Medical College, Chengde, 067000, P.R. China
| | - Lei Zheng
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, P.R. China
| | - Ping Wang
- Department of Respiratory, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Yaling Tian
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, P.R. China
| | - Shufeng Xu
- Department of Respiratory, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, P.R. China.
| |
Collapse
|
41
|
Sharma V, Goessling LS, Brar AK, Joshi CS, Mysorekar IU, Eghtesady P. Coxsackievirus B3 Infection Early in Pregnancy Induces Congenital Heart Defects Through Suppression of Fetal Cardiomyocyte Proliferation. J Am Heart Assoc 2021; 10:e017995. [PMID: 33440998 PMCID: PMC7955305 DOI: 10.1161/jaha.120.017995] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
Background Coxsackievirus B (CVB) is the most common cause of viral myocarditis. It targets cardiomyocytes through coxsackie and adenovirus receptor, which is highly expressed in the fetal heart. We hypothesized CVB3 can precipitate congenital heart defects when fetal infection occurs during critical window of gestation. Methods and Results We infected C57Bl/6 pregnant mice with CVB3 during time points in early gestation (embryonic day [E] 5, E7, E9, and E11). We used different viral titers to examine possible dose-response relationship and assessed viral loads in various fetal organs. Provided viral exposure occurred between E7 and E9, we observed characteristic features of ventricular septal defect (33.6%), abnormal myocardial architecture resembling noncompaction (23.5%), and double-outlet right ventricle (4.4%) among 209 viable fetuses examined. We observed a direct relationship between viral titers and severity of congenital heart defects, with apparent predominance among female fetuses. Infected dams remained healthy; we did not observe any maternal heart or placental injury suggestive of direct viral effects on developing heart as likely cause of congenital heart defects. We examined signaling pathways in CVB3-exposed hearts using RNA sequencing, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and immunohistochemistry. Signaling proteins of the Hippo, tight junction, transforming growth factor-β1, and extracellular matrix proteins were the most highly enriched in CVB3-infected fetuses with ventricular septal defects. Moreover, cardiomyocyte proliferation was 50% lower in fetuses with ventricular septal defects compared with uninfected controls. Conclusions We conclude prenatal CVB3 infection induces congenital heart defects. Alterations in myocardial proliferate capacity and consequent changes in cardiac architecture and trabeculation appear to account for most of observed phenotypes.
Collapse
Affiliation(s)
- Vipul Sharma
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Lisa S. Goessling
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Anoop K. Brar
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| | - Chetanchandra S. Joshi
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMO
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMO
| | - Indira U. Mysorekar
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMO
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMO
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic SurgeryDepartment of SurgeryWashington University School of MedicineSt. LouisMO
| |
Collapse
|
42
|
The effect of acute heat stress on the innate immune function of rainbow trout based on the transcriptome. J Therm Biol 2021; 96:102834. [PMID: 33627272 DOI: 10.1016/j.jtherbio.2021.102834] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/09/2020] [Accepted: 01/02/2021] [Indexed: 01/21/2023]
Abstract
Heat stress is a condition in which the body's homeostasis is disturbed as a result of the rise in water temperature, resulting in the decline or even death of growth, immunity, and other functions. The mechanisms directing this response are not fully understood. To better characterize the effects of acute heat stress on the innate immune function of rainbow trout, we identified differentially regulated messenger RNA (mRNA) and non-coding RNA (ncRNA) in rainbow trout exposed to acute heat stress. Next-generation RNA sequencing and comprehensive bioinformatics analysis were conducted to characterize the transcriptome profiles, including mRNA, microRNA (miRNA), and long non-coding RNA (lncRNA). The head kidney of rainbow trout were exposed to acute heat stress at 22.5 °C for 24 h. A total of 2605 lncRNAs, 214 miRNAs, and 5608 mRNAs were identified as differentially regulated. Among these expressed genes differentially, 45 lncRNAs and 2 target genes, as well as 38 miRNAs and 14 target genes were significantly enriched in the innate immune response of rainbow trout. LncRNA is used as competitive endogenous RNA (ceRNA) to construct the ceRNA-miRNA-mRNA interaction network. Enrichment analysis of the Kyoto encyclopedia of genes and genomes (KEGG) of ceRNA, the differentially expressed genes related to the innate immune function of rainbow trout, were significantly enriched in the signaling pathway mediated by mitogen-activated protein kinase (MAPK). Overall, these analyses showed the effects of heat stress on the innate immune function in rainbow trout at the transcriptome level, providing a theoretical basis to improve the production and breeding of rainbow trout and the selection of new heat-resistant varieties.
Collapse
|
43
|
Abstract
The phenotypic trait of high bone mass (HBM) is an excellent example of the nexus between common and rare disease genetics. HBM may arise from carriage of many 'high bone mineral density [BMD]'-associated alleles, and certainly the genetic architecture of individuals with HBM is enriched with high BMD variants identified through genome-wide association studies of BMD. HBM may also arise as a monogenic skeletal disorder, due to abnormalities in bone formation, bone resorption, and/or bone turnover. Individuals with monogenic disorders of HBM usually, though not invariably, have other skeletal abnormalities (such as mandible enlargement) and thus are best regarded as having a skeletal dysplasia rather than just isolated high BMD. A binary etiological division of HBM into polygenic vs. monogenic, however, would be excessively simplistic: the phenotype of individuals carrying rare variants of large effect can still be modified by their common variant polygenic background, and by the environment. HBM disorders-whether predominantly polygenic or monogenic in origin-are not only interesting clinically and genetically: they provide insights into bone processes that can be exploited therapeutically, with benefits both for individuals with these rare bone disorders and importantly for the many people affected by the commonest bone disease worldwide-i.e., osteoporosis. In this review we detail the genetic architecture of HBM; we provide a conceptual framework for considering HBM in the clinical context; and we discuss monogenic and polygenic causes of HBM with particular emphasis on anabolic causes of HBM.
Collapse
Affiliation(s)
- Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
44
|
Influence of the TGF-β Superfamily on Osteoclasts/Osteoblasts Balance in Physiological and Pathological Bone Conditions. Int J Mol Sci 2020; 21:ijms21207597. [PMID: 33066607 PMCID: PMC7589189 DOI: 10.3390/ijms21207597] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
The balance between bone forming cells (osteoblasts/osteocytes) and bone resorbing cells (osteoclasts) plays a crucial role in tissue homeostasis and bone repair. Several hormones, cytokines, and growth factors-in particular the members of the TGF-β superfamily such as the bone morphogenetic proteins-not only regulate the proliferation, differentiation, and functioning of these cells, but also coordinate the communication between them to ensure an appropriate response. Therefore, this review focuses on TGF-β superfamily and its influence on bone formation and repair, through the regulation of osteoclastogenesis, osteogenic differentiation of stem cells, and osteoblasts/osteoclasts balance. After introducing the main types of bone cells, their differentiation and cooperation during bone remodeling and fracture healing processes are discussed. Then, the TGF-β superfamily, its signaling via canonical and non-canonical pathways, as well as its regulation by Wnt/Notch or microRNAs are described and discussed. Its important role in bone homeostasis, repair, or disease is also highlighted. Finally, the clinical therapeutic uses of members of the TGF-β superfamily and their associated complications are debated.
Collapse
|
45
|
Kaliya-Perumal AK, Carney TJ, Ingham PW. Fibrodysplasia ossificans progressiva: current concepts from bench to bedside. Dis Model Mech 2020; 13:13/9/dmm046441. [PMID: 32988985 PMCID: PMC7522019 DOI: 10.1242/dmm.046441] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heterotopic ossification (HO) is a disorder characterised by the formation of ectopic bone in soft tissue. Acquired HO typically occurs in response to trauma and is relatively common, yet its aetiology remains poorly understood. Genetic forms, by contrast, are very rare, but provide insights into the mechanisms of HO pathobiology. Fibrodysplasia ossificans progressiva (FOP) is the most debilitating form of HO. All patients reported to date carry heterozygous gain-of-function mutations in the gene encoding activin A receptor type I (ACVR1). These mutations cause dysregulated bone morphogenetic protein (BMP) signalling, leading to HO at extraskeletal sites including, but not limited to, muscles, ligaments, tendons and fascia. Ever since the identification of the causative gene, developing a cure for FOP has been a focus of investigation, and studies have decoded the pathophysiology at the molecular and cellular levels, and explored novel management strategies. Based on the established role of BMP signalling throughout HO in FOP, therapeutic modalities that target multiple levels of the signalling cascade have been designed, and some drugs have entered clinical trials, holding out hope of a cure. A potential role of other signalling pathways that could influence the dysregulated BMP signalling and present alternative therapeutic targets remains a matter of debate. Here, we review the recent FOP literature, including pathophysiology, clinical aspects, animal models and current management strategies. We also consider how this research can inform our understanding of other types of HO and highlight some of the remaining knowledge gaps. Summary: Fibrodysplasia ossificans progressiva is a rare disease characterised by progressive heterotopic bone formation. Here, we present a comprehensive summary of the recent literature on this debilitating condition and discuss approaches to solving this clinical puzzle.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore
| | - Tom J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos 138673, Singapore
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore .,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos 138673, Singapore
| |
Collapse
|
46
|
Rajapakse D, Peterson K, Mishra S, Fan J, Lerner J, Campos M, Wistow G. Amelotin is expressed in retinal pigment epithelium and localizes to hydroxyapatite deposits in dry age-related macular degeneration. Transl Res 2020; 219:45-62. [PMID: 32160961 PMCID: PMC7197213 DOI: 10.1016/j.trsl.2020.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/28/2022]
Abstract
Deposition of hydroxyapatite (HAP) basal to the retinal pigment epithelium (RPE) is linked to the progression of age-related macular degeneration (AMD). Serum-deprivation of RPE cells in culture mimics some features of AMD. We now show that serum-deprivation also leads to the induction of amelotin (AMTN), a protein involved in hydroxyapatite mineralization in enamel. HAP is formed in our culture model and is blocked by siRNA inhibition of AMTN expression. In situ hybridization and immunofluorescence imaging of human eye tissue show that AMTN is expressed in RPE of donor eyes with geographic atrophy ("dry" AMD) in regions with soft drusen containing HAP spherules or nodules. AMTN is not found in hard drusen, normal RPE, or donor eyes diagnosed with wet AMD. These findings suggest that AMTN is involved in formation of HAP spherules or nodules in AMD, and as such provides a new therapeutic target for slowing disease progression.
Collapse
Affiliation(s)
- Dinusha Rajapakse
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine Peterson
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanghamitra Mishra
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Jianguo Fan
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Joshua Lerner
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Campos
- Histopathology Core Facility, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
47
|
Wang JJ, Zhang T, Chen QM, Zhang RQ, Li L, Cheng SF, Shen W, Lei CZ. Genomic Signatures of Selection Associated With Litter Size Trait in Jining Gray Goat. Front Genet 2020; 11:286. [PMID: 32273886 PMCID: PMC7113370 DOI: 10.3389/fgene.2020.00286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/09/2020] [Indexed: 12/28/2022] Open
Abstract
Litter size (LS), an important economic trait in livestock, is so complicate that involves many aspects of reproduction, the underlying mechanism of which particularly in goat has always been scanty. To uncover the genetic basis of LS, the genomic sequence of Jining Gray goat groups (one famous breed for high prolificacy in China) with LS 1, 2, and 3 for firstborn was analyzed, obtaining 563.67 Gb sequence data and a total of 31,864,651 high-quality single nucleotide polymorphisms loci were identified. Particularly, the increased heterozygosity in higher LS groups, and large continuous homozygous segments associated with lower LS group had been uncovered. Through an integrated analysis of three popular methods for detecting selective sweeps (Fst, nucleotide diversity, and Tajima’s D statistic), 111 selected regions and 42 genes associated with LS were scanned genome wide. The candidate genes with highest selective signatures included KIT, KCNH7, and KMT2E in LS2 and PAK1, PRKAA1, and SMAD9 in LS3 group, respectively. Meanwhile, functional terms of programmed cell death involved in cell development and regulation of insulin receptor signaling pathway were mostly enriched with 42 candidate genes, which also included reproduction related terms of steroid metabolic process and cellular response to hormone stimulus. In conclusion, our study identified novel candidate genes involving in regulation of LS in goat, which expand our understanding of genetic fundament of reproductive ability, and the novel insights regarding to LS would be potentially applied to improve reproductive performance.
Collapse
Affiliation(s)
- Jun-Jie Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Teng Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Qiu-Ming Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rui-Qian Zhang
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lan Li
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Shun-Feng Cheng
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Chu-Zhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
48
|
Gregson CL, Bergen DJM, Leo P, Sessions RB, Wheeler L, Hartley A, Youlten S, Croucher PI, McInerney‐Leo AM, Fraser W, Tang JCY, Anderson L, Marshall M, Sergot L, Paternoster L, Davey Smith G, The AOGC Consortium, Brown MA, Hammond C, Kemp JP, Tobias JH, Duncan EL. A Rare Mutation in SMAD9 Associated With High Bone Mass Identifies the SMAD-Dependent BMP Signaling Pathway as a Potential Anabolic Target for Osteoporosis. J Bone Miner Res 2020; 35:92-105. [PMID: 31525280 PMCID: PMC7004081 DOI: 10.1002/jbmr.3875] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/19/2019] [Accepted: 08/25/2019] [Indexed: 01/17/2023]
Abstract
Novel anabolic drug targets are needed to treat osteoporosis. Having established a large national cohort with unexplained high bone mass (HBM), we aimed to identify a novel monogenic cause of HBM and provide insight into a regulatory pathway potentially amenable to therapeutic intervention. We investigated a pedigree with unexplained HBM in whom previous sequencing had excluded known causes of monogenic HBM. Whole exome sequencing identified a rare (minor allele frequency 0.0023), highly evolutionarily conserved missense mutation in SMAD9 (c.65T>C, p.Leu22Pro) segregating with HBM in this autosomal dominant family. The same mutation was identified in another two unrelated individuals both with HBM. In silico protein modeling predicts the mutation severely disrupts the MH1 DNA-binding domain of SMAD9. Affected individuals have bone mineral density (BMD) Z-scores +3 to +5, mandible enlargement, a broad frame, torus palatinus/mandibularis, pes planus, increased shoe size, and a tendency to sink when swimming. Peripheral quantitative computed tomography (pQCT) measurement demonstrates increased trabecular volumetric BMD and increased cortical thickness conferring greater predicted bone strength; bone turnover markers are low/normal. Notably, fractures and nerve compression are not found. Both genome-wide and gene-based association testing involving estimated BMD measured at the heel in 362,924 white British subjects from the UK Biobank Study showed strong associations with SMAD9 (PGWAS = 6 × 10-16 ; PGENE = 8 × 10-17 ). Furthermore, we found Smad9 to be highly expressed in both murine cortical bone-derived osteocytes and skeletal elements of zebrafish larvae. Our findings support SMAD9 as a novel HBM gene and a potential novel osteoanabolic target for osteoporosis therapeutics. SMAD9 is thought to inhibit bone morphogenetic protein (BMP)-dependent target gene transcription to reduce osteoblast activity. Thus, we hypothesize SMAD9 c.65T>C is a loss-of-function mutation reducing BMP inhibition. Lowering SMAD9 as a potential novel anabolic mechanism for osteoporosis therapeutics warrants further investigation. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Celia L Gregson
- Musculoskeletal Research Unit, Translational Health SciencesBristol Medical School, University of BristolBristolUK
| | - Dylan J. M. Bergen
- Musculoskeletal Research Unit, Translational Health SciencesBristol Medical School, University of BristolBristolUK
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life SciencesUniversity of BristolBristolUK
| | - Paul Leo
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Richard B Sessions
- Faculty of Life SciencesSchool of Biochemistry, University of BristolBristolUK
| | - Lawrie Wheeler
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - April Hartley
- Musculoskeletal Research Unit, Translational Health SciencesBristol Medical School, University of BristolBristolUK
- Medical Research Council Integrative Epidemiology Unit, Population Health SciencesBristol Medical School, University of BristolBristolUK
| | - Scott Youlten
- Division of Bone BiologyGarvan Institute of Medical ResearchSydneyAustralia
| | - Peter I Croucher
- Division of Bone BiologyGarvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical School, UNSW SydneySydneyAustralia
- School of Biotechnology and Biomolecular Sciences, UNSW SydneySydneyAustralia
| | - Aideen M McInerney‐Leo
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina InstituteBrisbaneAustralia
| | - William Fraser
- Norwich Medical School, University of East AngliaNorwichUK
- Department of DiabetesEndocrinology and Clinical Biochemistry, Norfolk and Norwich University Hospital NHS Foundation TrustNorwichUK
| | | | - Lisa Anderson
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Mhairi Marshall
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Leon Sergot
- Severn School of Radiology, Severn DeaneryBristolUK
| | - Lavinia Paternoster
- Medical Research Council Integrative Epidemiology Unit, Population Health SciencesBristol Medical School, University of BristolBristolUK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, Population Health SciencesBristol Medical School, University of BristolBristolUK
| | - The AOGC Consortium
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Matthew A Brown
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Chrissy Hammond
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life SciencesUniversity of BristolBristolUK
| | - John P Kemp
- Medical Research Council Integrative Epidemiology Unit, Population Health SciencesBristol Medical School, University of BristolBristolUK
- Faculty of MedicineThe University of Queensland Diamantina Institute, The University of QueenslandWoolloongabbaAustralia
| | - Jon H Tobias
- Musculoskeletal Research Unit, Translational Health SciencesBristol Medical School, University of BristolBristolUK
| | - Emma L Duncan
- Faculty of Health, Translational Genomics GroupInstitute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
- Department of Endocrinology and DiabetesRoyal Brisbane & Women's HospitalHerstonAustralia
- Faculty of MedicineUniversity of QueenslandHerstonAustralia
| |
Collapse
|
49
|
Hui Y, Wei PJ, Xia J, Wang YT, Zheng CH. MECoRank: cancer driver genes discovery simultaneously evaluating the impact of SNVs and differential expression on transcriptional networks. BMC Med Genomics 2019; 12:140. [PMID: 31888623 PMCID: PMC6936061 DOI: 10.1186/s12920-019-0582-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/10/2019] [Indexed: 01/09/2023] Open
Abstract
Background Although there are huge volumes of genomic data, how to decipher them and identify driver events is still a challenge. The current methods based on network typically use the relationship between genomic events and consequent changes in gene expression to nominate putative driver genes. But there may exist some relationships within the transcriptional network. Methods We developed MECoRank, a novel method that improves the recognition accuracy of driver genes. MECoRank is based on bipartite graph to propagates the scores via an iterative process. After iteration, we will obtain a ranked gene list for each patient sample. Then, we applied the Condorcet voting method to determine the most impactful drivers in a population. Results We applied MECoRank to three cancer datasets to reveal candidate driver genes which have a greater impact on gene expression. Experimental results show that our method not only can identify more driver genes that have been validated than other methods, but also can recognize some impactful novel genes which have been proved to be more important in literature. Conclusions We propose a novel approach named MECoRank to prioritize driver genes based on their impact on the expression in the molecular interaction network. This method not only assesses mutation’s effect on the transcriptional network, but also assesses the differential expression’s effect within the transcriptional network. And the results demonstrated that MECoRank has better performance than the other competing approaches in identifying driver genes.
Collapse
Affiliation(s)
- Ying Hui
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Pi-Jing Wei
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Junfeng Xia
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Yu-Tian Wang
- School of Software Engineering, Qufu Normal University, Qufu, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China.
| |
Collapse
|
50
|
Wang CY, Xiao X, Bayer A, Xu Y, Dev S, Canali S, Nair AV, Masia R, Babitt JL. Ablation of Hepatocyte Smad1, Smad5, and Smad8 Causes Severe Tissue Iron Loading and Liver Fibrosis in Mice. Hepatology 2019; 70:1986-2002. [PMID: 31127639 PMCID: PMC6874904 DOI: 10.1002/hep.30780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Abstract
A failure of iron to appropriately regulate liver hepcidin production is central to the pathogenesis of hereditary hemochromatosis. SMAD1/5 transcription factors, activated by bone morphogenetic protein (BMP) signaling, are major regulators of hepcidin production in response to iron; however, the role of SMAD8 and the contribution of SMADs to hepcidin production by other systemic cues remain uncertain. Here, we generated hepatocyte Smad8 single (Smad8fl/fl ;Alb-Cre+ ), Smad1/5/8 triple (Smad158;Alb-Cre+ ), and littermate Smad1/5 double (Smad15;Alb-Cre+ ) knockout mice to investigate the role of SMAD8 in hepcidin and iron homeostasis regulation and liver injury. We found that Smad8;Alb-Cre+ mice exhibited no iron phenotype, whereas Smad158;Alb-Cre+ mice had greater iron overload than Smad15;Alb-Cre+ mice. In contrast to the sexual dimorphism reported for wild-type mice and other hemochromatosis models, hepcidin deficiency and extrahepatic iron loading were similarly severe in Smad15;Alb-Cre+ and Smad158;Alb-Cre+ female compared with male mice. Moreover, epidermal growth factor (EGF) failed to suppress hepcidin in Smad15;Alb-Cre+ hepatocytes. Conversely, hepcidin was still increased by lipopolysaccharide in Smad158;Alb-Cre+ mice, although lower basal hepcidin resulted in lower maximal hepcidin. Finally, unlike most mouse hemochromatosis models, Smad158;Alb-Cre+ developed liver injury and fibrosis at 8 weeks. Liver injury and fibrosis were prevented in Smad158;Alb-Cre+ mice by a low-iron diet and were minimal in iron-loaded Cre- mice. Conclusion: Hepatocyte Smad1/5/8 knockout mice are a model of hemochromatosis that encompasses liver injury and fibrosis seen in human disease. These mice reveal the redundant but critical role of SMAD8 in hepcidin and iron homeostasis regulation, establish a requirement for SMAD1/5/8 in hepcidin regulation by testosterone and EGF but not inflammation, and suggest a pathogenic role for both iron loading and SMAD1/5/8 deficiency in liver injury and fibrosis.
Collapse
Affiliation(s)
- Chia-Yu Wang
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology,Address correspondence to: Chia-Yu Wang, Massachusetts General Hospital, 185 Cambridge St., CPZN-8150, Boston, MA 02114, Phone: (617)-724-9078, Fax: (617)-643-3182,
| | - Xia Xiao
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Abraham Bayer
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Yang Xu
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Som Dev
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Susanna Canali
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Anil V. Nair
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Ricard Masia
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jodie L. Babitt
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| |
Collapse
|