1
|
Passamonti MM, Milanesi M, Cattaneo L, Ramirez-Diaz J, Stella A, Barbato M, Braz CU, Negrini R, Giannuzzi D, Pegolo S, Cecchinato A, Trevisi E, Williams JL, Ajmone Marsan P. Unraveling metabolic stress response in dairy cows: Genetic control of plasma biomarkers throughout lactation and the transition period. J Dairy Sci 2024; 107:9602-9614. [PMID: 38945260 DOI: 10.3168/jds.2023-24630] [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: 12/29/2023] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
Breeding animals able to effectively respond to stress could be a long-term, sustainable, and affordable strategy to improve resilience and welfare in livestock systems. In the present study, the concentrations of 29 plasma biomarkers were used as candidate endophenotypes for metabolic stress response in single-SNP, gene- and haplotype-based GWAS using 739 healthy lactating Italian Holstein cows and 88,271 variants. Significant genetic associations were found in all the 3 GWAS approaches for plasma γ-glutamyl transferase concentration on BTA17, for paraoxonase on BTA4, and for alkaline phosphatase and zinc on BTA2. On these chromosomes, single-SNP and gene-based chromosome-wide association studies were performed, confirming GWAS findings. The signals identified for paraoxonase, γ-glutamyl transferase, and alkaline phosphatase were in proximity to the genes coding for them. The heritability of these 4 biomarkers ranged from moderate to high (from 0.39 to 0.54). Plasma biomarkers are known to undergo large changes in concentration during metabolic stress in the transition period, with an interindividual variability in the rate of change and recovery time. Genetics may account in part for these differences. To assess this, we studied a subset of 139 periparturient cows homozygous at 3 SNPs known to be respectively associated with concentration of plasma ceruloplasmin, paraoxonase, and γ-glutamyl transferase. We compared the immune-metabolic profile measured in plasma at -7, +5, and +30 d relative to calving between groups of opposite homozygotes. A significant effect of the genotype was found on paraoxonase and γ-glutamyl transferase plasma concentration at all the 3 time points. No evidence for genotype effect was detected for ceruloplasmin. Understanding the genetic control underlying metabolic stress response may suggest new approaches to foster resilience in dairy cows.
Collapse
Affiliation(s)
- M M Passamonti
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - M Milanesi
- Department for Innovation in Biological, Agro-food and Forest Systems-DIBAF, Università della Tuscia, 01100 Viterbo, Italy
| | - L Cattaneo
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - J Ramirez-Diaz
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, 26900 Lodi, Italy
| | - A Stella
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, 26900 Lodi, Italy
| | - M Barbato
- Department of Veterinary Sciences, Università degli Studi di Messina, 98168 Messina, Italy
| | - C U Braz
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - R Negrini
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - D Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - S Pegolo
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - A Cecchinato
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - E Trevisi
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; Romeo and Enrica Invernizzi Research Center on Sustainable Dairy Production-CREI, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - J L Williams
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - P Ajmone Marsan
- Department of Animal Science, Food and Nutrition-DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; Romeo and Enrica Invernizzi Research Center on Sustainable Dairy Production-CREI, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy.
| |
Collapse
|
2
|
Zhao Z, Fan C, Wang S, Wang H, Deng H, Zeng S, Tang S, Li L, Xiong Z, Qiu X. Single-nucleus RNA and multiomics in situ pairwise sequencing reveals cellular heterogeneity of the abnormal ligamentum teres in patients with developmental dysplasia of the hip. Heliyon 2024; 10:e27803. [PMID: 38524543 PMCID: PMC10958365 DOI: 10.1016/j.heliyon.2024.e27803] [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: 09/02/2023] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Developmental dysplasia of the hip (DDH) is the most common hip deformity in pediatric orthopedics. One of the common pathological changes in DDH is the thickening and hypertrophy of the ligamentum teres. However, the underlying pathogenic mechanism responsible for these changes remains unclear. This study represents the first time that the heterogeneity of cell subsets in the abnormal ligamentum teres of patients with DDH has been resolved at the single-cell and spatial levels by snRNA-Seq and MiP-Seq. Through gene set enrichment and intercellular communication network analyses, we found that receptor-like cells and ligament stem cells may play an essential role in the pathological changes resulting in ligamentum teres thickening and hypertrophy. Eight ligand-receptor pairs related to the ECM-receptor pathway were observed to be closely associated with DDH. Further, using the Monocle R package, we predicted a differentiation trajectory of pericytes into two branches, leading to junctional ligament stem cells or fibroblasts. The expression of extracellular matrix-related genes along pseudotemporal trajectories was also investigated. Using MiP-Seq, we determined the expression distribution of marker genes specific to different cell types within the ligamentum teres, as well as differentially expressed DDH-associated genes at the spatial level.
Collapse
Affiliation(s)
- Zhenhui Zhao
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
- China Medical University, Shenyang, Liaoning Province, China
| | - Chuiqin Fan
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
- China Medical University, Shenyang, Liaoning Province, China
| | - Shiyou Wang
- Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Haoyu Wang
- Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Hansheng Deng
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Shuaidan Zeng
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Shengping Tang
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Li Li
- Shenzhen Luohu Maternity and Child Healthcare Hospital, Shenzhen, Guangdong Province, China
| | - Zhu Xiong
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
- China Medical University, Shenyang, Liaoning Province, China
| | - Xin Qiu
- Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| |
Collapse
|
3
|
Eom YJ, Kim JW, Rim YA, Lim J, Jung SI, Ju JH. Effects of stepwise administration of osteoprotegerin and parathyroid hormone-related peptide DNA vectors on bone formation in ovariectomized rat model. Sci Rep 2024; 14:2477. [PMID: 38291053 PMCID: PMC10827729 DOI: 10.1038/s41598-024-51957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Osteoporosis is a metabolic bone disease that impairs bone mineral density, microarchitecture, and strength. It requires continuous management, and further research into new treatment options is necessary. Osteoprotegerin (OPG) inhibits bone resorption and osteoclast activity. The objective of this study was to investigate the effects of stepwise administration of OPG-encoded minicircles (mcOPG) and a bone formation regulator, parathyroid hormone-related peptide (PTHrP)-encoded minicircles (mcPTHrP) in osteoporosis. The combined treatment with mcOPG and mcPTHrP significantly increased osteogenic marker expression in osteoblast differentiation compared with the single treatment groups. A model of postmenopausal osteoporosis was established in 12-week-old female rats through ovariectomy (OVX). After 8 weeks of OVX, mcOPG (80 µg/kg) was administered via intravenous injection. After 16 weeks of OVX, mcPTHrP (80 µg/kg) was injected once a week for 3 weeks. The bone microstructure in the femur was evaluated 24 weeks after OVX using micro-CT. In a proof-of-concept study, stepwise treatment with mcOPG and mcPTHrP on an OVX rat model significantly improved bone microstructure compared to treatment with mcOPG or mcPTHrP alone. These results suggest that stepwise treatment with mcOPG and mcPTHrP may be a potential treatment for osteoporosis.
Collapse
Affiliation(s)
- Ye Ji Eom
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jang-Woon Kim
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Jooyoung Lim
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Se In Jung
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center (CiRC), CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Doolittle ML, Khosla S, Saul D. Single-Cell Integration of BMD GWAS Results Prioritize Candidate Genes Influencing Age-Related Bone Loss. JBMR Plus 2023; 7:e10795. [PMID: 37808401 PMCID: PMC10556272 DOI: 10.1002/jbm4.10795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/17/2023] [Accepted: 06/19/2023] [Indexed: 10/10/2023] Open
Abstract
The regulation of bone mineral density (BMD) is highly influenced by genetics and age. Although genome-wide association studies (GWAS) for BMD have uncovered many genes through their proximity to associated variants (variant nearest-neighbor [VNN] genes), the cell-specific mechanisms of each VNN gene remain unclear. This is primarily due to the inability to prioritize these genes by cell type and age-related expression. Using age-related transcriptomics, we found that the expression of many VNN genes was upregulated in the bone and marrow from aged mice. Candidate genes from GWAS were investigated using single-cell RNA-sequencing (scRNA-seq) datasets to enrich for cell-specific expression signatures. VNN candidate genes are highly enriched in osteo-lineage cells, osteocytes, hypertrophic chondrocytes, and Lepr+ mesenchymal stem cells. These data were used to generate a "blueprint" for Cre-loxp mouse line selection for functional validation of candidate genes and further investigation of their role in BMD maintenance throughout aging. In VNN-gene-enriched cells, Sparc, encoding the extracellular matrix (ECM) protein osteonectin, was robustly expressed. This, along with expression of numerous other ECM genes, indicates that many VNN genes likely have roles in ECM deposition by osteoblasts. Overall, we provide data supporting streamlined translation of GWAS candidate genes to potential novel therapeutic targets for the treatment of osteoporosis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Madison L. Doolittle
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Sundeep Khosla
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Dominik Saul
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department for Trauma and Reconstructive SurgeryBG Clinic, University of TuebingenTuebingenGermany
| |
Collapse
|
5
|
Zhou M, Cui Y, Zuo S, Peng Q, Liu Y, Li X, Yang Y, He Q, Yu X, Zhou J, He Z, He Q. ZBTB40 is a telomere-associated protein and protects telomeres in human ALT cells. J Biol Chem 2023; 299:105053. [PMID: 37454741 PMCID: PMC10480536 DOI: 10.1016/j.jbc.2023.105053] [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: 10/22/2022] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
Alternative lengthening of telomeres (ALTs) mechanism is activated in some somatic, germ cells, and human cancer cells. However, the key regulators and mechanisms of the ALT pathway remain elusive. Here we demonstrated that ZBTB40 is a novel telomere-associated protein and binds to telomeric dsDNA through its N-terminal BTB (BR-C, ttk and bab) or POZ (Pox virus and Zinc finger) domain in ALT cells. Notably, the knockout or knockdown of ZBTB40 resulted in the telomere dysfunction-induced foci and telomere lengthening in the ALT cells. The results also show that ZBTB40 is associated with ALT-associated promyelocytic leukemia nuclear bodies, and the loss of ZBTB40 induces the accumulation of the ALT-associated promyelocytic leukemia nuclear bodies in U2OS cells. Taken together, our results implicate that ZBTB40 is a key player of telomere protection and telomere lengthening regulation in human ALT cells.
Collapse
Affiliation(s)
- Mingqing Zhou
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yinghong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Shanru Zuo
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Qiyao Peng
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yucong Liu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Xueguang Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yide Yang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Quanze He
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xing Yu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Junhua Zhou
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China.
| | - Quanyuan He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China.
| |
Collapse
|
6
|
Koike Y, Takahata M, Nakajima M, Otomo N, Suetsugu H, Liu X, Endo T, Imagama S, Kobayashi K, Kaito T, Kato S, Kawaguchi Y, Kanayama M, Sakai H, Tsuji T, Miyamoto T, Inose H, Yoshii T, Kashii M, Nakashima H, Ando K, Taniguchi Y, Takeuchi K, Ito S, Tomizuka K, Hikino K, Iwasaki Y, Kamatani Y, Maeda S, Nakajima H, Mori K, Seichi A, Fujibayashi S, Kanchiku T, Watanabe K, Tanaka T, Kida K, Kobayashi S, Takahashi M, Yamada K, Takuwa H, Lu HF, Niida S, Ozaki K, Momozawa Y, Yamazaki M, Okawa A, Matsumoto M, Iwasaki N, Terao C, Ikegawa S. Genetic insights into ossification of the posterior longitudinal ligament of the spine. eLife 2023; 12:e86514. [PMID: 37461309 DOI: 10.7554/elife.86514] [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: 01/30/2023] [Accepted: 05/22/2023] [Indexed: 07/20/2023] Open
Abstract
Ossification of the posterior longitudinal ligament of the spine (OPLL) is an intractable disease leading to severe neurological deficits. Its etiology and pathogenesis are primarily unknown. The relationship between OPLL and comorbidities, especially type 2 diabetes (T2D) and high body mass index (BMI), has been the focus of attention; however, no trait has been proven to have a causal relationship. We conducted a meta-analysis of genome-wide association studies (GWASs) using 22,016 Japanese individuals and identified 14 significant loci, 8 of which were previously unreported. We then conducted a gene-based association analysis and a transcriptome-wide Mendelian randomization approach and identified three candidate genes for each. Partitioning heritability enrichment analyses observed significant enrichment of the polygenic signals in the active enhancers of the connective/bone cell group, especially H3K27ac in chondrogenic differentiation cells, as well as the immune/hematopoietic cell group. Single-cell RNA sequencing of Achilles tendon cells from a mouse Achilles tendon ossification model confirmed the expression of genes in GWAS and post-GWAS analyses in mesenchymal and immune cells. Genetic correlations with 96 complex traits showed positive correlations with T2D and BMI and a negative correlation with cerebral aneurysm. Mendelian randomization analysis demonstrated a significant causal effect of increased BMI and high bone mineral density on OPLL. We evaluated the clinical images in detail and classified OPLL into cervical, thoracic, and the other types. GWAS subanalyses identified subtype-specific signals. A polygenic risk score for BMI demonstrated that the effect of BMI was particularly strong in thoracic OPLL. Our study provides genetic insight into the etiology and pathogenesis of OPLL and is expected to serve as a basis for future treatment development.
Collapse
Affiliation(s)
- Yoshinao Koike
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Takahata
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiro Nakajima
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Nao Otomo
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Nagoya, Japan
| | - Hiroyuki Suetsugu
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shiro Imagama
- Department of Orthopedics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyoshi Kobayashi
- Department of Orthopedics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kato
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | - Masahiro Kanayama
- Department of Orthopedics, Hakodate Central General Hospital, Hakodate, Japan
| | - Hiroaki Sakai
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takashi Tsuji
- Department of Orthopedic Surgery, Keio University School of Medicine, Nagoya, Japan
- Department of Spine and Spinal Cord Surgery, Fujita Health University, Toyoake, Japan
| | - Takeshi Miyamoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Inose
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshitaka Yoshii
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masafumi Kashii
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroaki Nakashima
- Department of Orthopedics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kei Ando
- Department of Orthopedics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Taniguchi
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Takeuchi
- Department of Orthopaedic Surgery, National Okayama Medical Center, Okayama, Japan
| | - Shuji Ito
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Keiko Hikino
- Laboratory for Pharmacogenomics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Yusuke Iwasaki
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Shingo Maeda
- Department of Bone and Joint Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hideaki Nakajima
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kanji Mori
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Atsushi Seichi
- Department of Orthopedics, Jichi Medical University, Shimotsuke, Japan
| | - Shunsuke Fujibayashi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsukasa Kanchiku
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Kei Watanabe
- Department of Orthopaedic Surgery, Niigata University Medical and Dental General Hospital, Nankoku, Japan
| | - Toshihiro Tanaka
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kazunobu Kida
- Department of Orthopaedic Surgery, Kochi Medical School, Nankoku, Japan
| | - Sho Kobayashi
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masahito Takahashi
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, Tokyo, Japan
| | - Kei Yamada
- Department of Orthopaedic Surgery, Kurume University School of Medicine, Obu, Japan
| | - Hiroshi Takuwa
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Hsing-Fang Lu
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Million-Person Precision Medicine Initiative, China Medical University Hospital, Taichung, Taiwan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Okawa
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Nagoya, Japan
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
7
|
Cui Y, Zhou M, He Q, He Z. Zbtb40 Deficiency Leads to Morphological and Phenotypic Abnormalities of Spermatocytes and Spermatozoa and Causes Male Infertility. Cells 2023; 12:cells12091264. [PMID: 37174664 PMCID: PMC10177581 DOI: 10.3390/cells12091264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Studies on the gene regulation of spermatogenesis are of unusual significance for maintaining male reproduction and treating male infertility. Here, we have demonstrated, for the first time, that a loss of ZBTB40 function leads to abnormalities in the morphological and phenotypic characteristics of mouse spermatocytes and spermatids as well as male infertility. We revealed that Zbtb40 was expressed in spermatocytes of mouse testes, and it was co-localized with γH2AX in mouse secondary spermatocytes. Interestingly, spermatocytes of Zbtb40 knockout mice had longer telomeres, compromised double-strand break (DSB) repair in the sex chromosome, and a higher apoptosis ratio compared to wild-type (WT) mice. The testis weight, testicular volume, and cauda epididymis body weight of the Zbtb40+/- male mice were significantly lower than in WT mice. Mating tests indicated that Zbtb40+/- male mice were able to mate normally, but they failed to produce any pups. Notably, sperm of Zbtb40+/- mice showed flagellum deformities and abnormal acrosome biogenesis. Furthermore, a ZBTB40 mutation was associated with non-obstructive azoospermia. Our results implicate that ZBTB40 deficiency leads to morphological and phenotypic abnormalities of spermatocytes and spermatids and causes male infertility. This study thus offers a new genetic mechanism regulating mammalian spermatogenesis and provides a novel target for gene therapy in male infertility.
Collapse
Affiliation(s)
- Yinghong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- The Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410013, China
- The Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Mingqing Zhou
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Quanyuan He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- The Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410013, China
- The Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
8
|
Lian WS, Wu RW, Ko JY, Chen YS, Wang SY, Jahr H, Wang FS. Inhibition of histone lysine demethylase 6A promotes chondrocytic activity and attenuates osteoarthritis development through repressing H3K27me3 enhancement of Wnt10a. Int J Biochem Cell Biol 2023; 158:106394. [PMID: 36871937 DOI: 10.1016/j.biocel.2023.106394] [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: 10/19/2022] [Revised: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Histone hypermethylation represses gene transcription, which affects cartilage homeostasis or joint remodeling. Trimethylation of lysine 27 of histone 3 (H3K27me3) changes epigenome signatures, regulating tissue metabolism. This study aimed to investigate whether loss of H3K27me3 demethylase Kdm6a function affected osteoarthritis development. We revealed that chondrocyte-specific Kdm6a knockout mice developed relatively long femurs and tibiae as compared to wild-type mice. Kdm6a deletion mitigated osteoarthritis symptoms, including articular cartilage loss, osteophyte formation, subchondral trabecular bone loss, and irregular walking patterns of destabilized medial meniscus-injured knees. In vitro, loss of Kdm6a function compromised the loss in expression of key chondrocyte markers Sox9, collagen II, and aggrecan and improved glycosaminoglycan production in inflamed chondrocytes. RNA sequencing showed that Kdm6a loss changed transcriptomic profiles, which contributed to histone signaling, NADPH oxidase, Wnt signaling, extracellular matrix, and cartilage development in articular cartilage. Chromatin immunoprecipitation sequencing uncovered that Kdm6a knockout affected H3K27me3 binding epigenome, repressing Wnt10a and Fzd10 transcription. Wnt10a was, among others, functional molecules regulated by Kdm6a. Forced Wnt10a expression attenuated Kdm6a deletion-induced glycosaminoglycan overproduction. Intra-articular administration with Kdm6a inhibitor GSK-J4 attenuated articular cartilage erosion, synovitis, and osteophyte formation, improving gait profiles of injured joints. In conclusion, Kdm6a loss promoted transcriptomic landscapes contributing to extracellular matrix synthesis and compromised epigenetic H3K27me3-mediated promotion of Wnt10a signaling, preserving chondrocytic activity to attenuate osteoarthritic degeneration. We highlighted the chondroprotective effects of Kdm6a inhibitor for mitigating the development of osteoarthritic disorders.
Collapse
Affiliation(s)
- Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, Germany; Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
9
|
Ackert-Bicknell C, Karasik D. Proceedings of the Post-Genome Analysis for Musculoskeletal Biology Workshop. Curr Osteoporos Rep 2023; 21:184-192. [PMID: 36869984 DOI: 10.1007/s11914-023-00781-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/05/2023]
Abstract
PURPOSE OF THE REVIEW Herein, we report on the proceedings of the workshop entitled "Post-Genome analysis for musculoskeletal biology" that was held in July of 2022 in Safed, Galilee, Israel. Supported by the Israel Science Foundation, the goal of this workshop was to bring together established investigators and their trainees who were interested in understanding the etiology of musculoskeletal disease, from Israel and from around the world. RECENT FINDINGS Presentations at this workshop spanned the spectrum from basic science to clinical studies. A major emphasis of the discussion centered on genetic studies in humans, and the limitations and advantages of such studies. The power of coupling studies using human data with functional follow-up studies in pre-clinical models such as mice, rats, and zebrafish was discussed in depth. The advantages and limitations of mice and zebrafish for faithfully modelling aspects of human disease were debated, specifically in the context of age-related diseases such as osteoporosis, osteoarthritis, adult-onset auto-immune disease, and osteosarcopenia. There remain significant gaps in our understanding of the nature and etiology of human musculoskeletal disease. While therapies and medications exist, much work is still needed to find safe and effective interventions for all patients suffering from diseases associated with age-related deterioration of musculoskeletal tissues. The potential of forward and reverse genetic studies has not been exhausted for diseases of muscles, joints, and bones.
Collapse
Affiliation(s)
- Cheryl Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado, Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO, 80045, USA.
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
10
|
Akcan TS, Vilov S, Heinig M. Predictive model of transcriptional elongation control identifies trans regulatory factors from chromatin signatures. Nucleic Acids Res 2023; 51:1608-1624. [PMID: 36727445 PMCID: PMC9976927 DOI: 10.1093/nar/gkac1272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/09/2022] [Accepted: 01/12/2023] [Indexed: 02/03/2023] Open
Abstract
Promoter-proximal Polymerase II (Pol II) pausing is a key rate-limiting step for gene expression. DNA and RNA-binding trans-acting factors regulating the extent of pausing have been identified. However, we lack a quantitative model of how interactions of these factors determine pausing, therefore the relative importance of implicated factors is unknown. Moreover, previously unknown regulators might exist. Here we address this gap with a machine learning model that accurately predicts the extent of promoter-proximal Pol II pausing from large-scale genome and transcriptome binding maps and gene annotation and sequence composition features. We demonstrate high accuracy and generalizability of the model by validation on an independent cell line which reveals the model's cell line agnostic character. Model interpretation in light of prior knowledge about molecular functions of regulatory factors confirms the interconnection of pausing with other RNA processing steps. Harnessing underlying feature contributions, we assess the relative importance of each factor, quantify their predictive effects and systematically identify previously unknown regulators of pausing. We additionally identify 16 previously unknown 7SK ncRNA interacting RNA-binding proteins predictive of pausing. Our work provides a framework to further our understanding of the regulation of the critical early steps in transcriptional elongation.
Collapse
Affiliation(s)
- Toray S Akcan
- Institute of Computational Biology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Department of Computer Science, TUM School of Computation, Information and Technology, Technical University Munich, Munich, Germany
| | - Sergey Vilov
- Institute of Computational Biology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Department of Computer Science, TUM School of Computation, Information and Technology, Technical University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Association, Partner Site Munich, 10785 Berlin, Germany
| |
Collapse
|
11
|
Cottone L, Ligammari L, Lee HM, Knowles HJ, Henderson S, Bianco S, Davies C, Strauss S, Amary F, Leite AP, Tirabosco R, Haendler K, Schultze JL, Herrero J, O’Donnell P, Grigoriadis AE, Salomoni P, Flanagan AM. Aberrant paracrine signalling for bone remodelling underlies the mutant histone-driven giant cell tumour of bone. Cell Death Differ 2022; 29:2459-2471. [PMID: 36138226 PMCID: PMC9750984 DOI: 10.1038/s41418-022-01031-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 01/31/2023] Open
Abstract
Oncohistones represent compelling evidence for a causative role of epigenetic perturbations in cancer. Giant cell tumours of bone (GCTs) are characterised by a mutated histone H3.3 as the sole genetic driver present in bone-forming osteoprogenitor cells but absent from abnormally large bone-resorbing osteoclasts which represent the hallmark of these neoplasms. While these striking features imply a pathogenic interaction between mesenchymal and myelomonocytic lineages during GCT development, the underlying mechanisms remain unknown. We show that the changes in the transcriptome and epigenome in the mesenchymal cells caused by the H3.3-G34W mutation contribute to increase osteoclast recruitment in part via reduced expression of the TGFβ-like soluble factor, SCUBE3. Transcriptional changes in SCUBE3 are associated with altered histone marks and H3.3G34W enrichment at its enhancer regions. In turn, osteoclasts secrete unregulated amounts of SEMA4D which enhances proliferation of mutated osteoprogenitors arresting their maturation. These findings provide a mechanism by which GCTs undergo differentiation in response to denosumab, a drug that depletes the tumour of osteoclasts. In contrast, hTERT alterations, commonly found in malignant GCT, result in the histone-mutated neoplastic cells being independent of osteoclasts for their proliferation, predicting unresponsiveness to denosumab. We provide a mechanism for the initiation of GCT, the basis of which is dysfunctional cross-talk between bone-forming and bone-resorbing cells. The findings highlight the role of tumour/microenvironment bidirectional interactions in tumorigenesis and how this is exploited in the treatment of GCT.
Collapse
Affiliation(s)
- Lucia Cottone
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Lorena Ligammari
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Hang-Mao Lee
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Helen J. Knowles
- grid.4991.50000 0004 1936 8948Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD UK
| | - Stephen Henderson
- grid.83440.3b0000000121901201Bill Lyons Informatics Centre (BLIC), UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Sara Bianco
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK ,grid.83440.3b0000000121901201Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Christopher Davies
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK ,grid.416177.20000 0004 0417 7890Department of Histopathology, Royal National Orthopaedic Hospital, Middlesex, Stanmore, HA7 4LP UK
| | - Sandra Strauss
- grid.439749.40000 0004 0612 2754London Sarcoma Service, University College London Hospitals Foundation Trust, London, WC1E 6DD UK
| | - Fernanda Amary
- grid.416177.20000 0004 0417 7890Department of Histopathology, Royal National Orthopaedic Hospital, Middlesex, Stanmore, HA7 4LP UK
| | - Ana Paula Leite
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK ,grid.83440.3b0000000121901201Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Roberto Tirabosco
- grid.416177.20000 0004 0417 7890Department of Histopathology, Royal National Orthopaedic Hospital, Middlesex, Stanmore, HA7 4LP UK
| | - Kristian Haendler
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany ,grid.10388.320000 0001 2240 3300Platform for Single Cell Genomics and Epigenomics (PRECISE) at the DZNE and the University of Bonn, 53127 Bonn, Germany ,grid.4562.50000 0001 0057 2672Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - Joachim L. Schultze
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany ,grid.10388.320000 0001 2240 3300Platform for Single Cell Genomics and Epigenomics (PRECISE) at the DZNE and the University of Bonn, 53127 Bonn, Germany ,grid.10388.320000 0001 2240 3300Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Javier Herrero
- grid.83440.3b0000000121901201Bill Lyons Informatics Centre (BLIC), UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Paul O’Donnell
- grid.416177.20000 0004 0417 7890Department of Radiology, Royal National Orthopaedic Hospital, Middlesex, Stanmore, HA7 4LP UK
| | - Agamemnon E. Grigoriadis
- grid.239826.40000 0004 0391 895XCentre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London, SE1 9RT UK
| | - Paolo Salomoni
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany ,grid.83440.3b0000000121901201Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6BT UK
| | - Adrienne M. Flanagan
- grid.83440.3b0000000121901201Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT UK ,grid.416177.20000 0004 0417 7890Department of Histopathology, Royal National Orthopaedic Hospital, Middlesex, Stanmore, HA7 4LP UK
| |
Collapse
|
12
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
13
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
14
|
Cai X, Zhou J, Deng J, Chen Z. Prognostic biomarker SMARCC1 and its association with immune infiltrates in hepatocellular carcinoma. Cancer Cell Int 2021; 21:701. [PMID: 34937564 PMCID: PMC8697473 DOI: 10.1186/s12935-021-02413-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Background Epigenetic alterations contribute greatly to metastasis and dissemination in hepatocellular carcinoma (HCC). SMARCC1, as a SWI/SNF chromatin remodeling factor, has been reported to play important roles in many cancers. For the first time, with the bioinformatics analysis and wet-bench experiments, we explored the biological significance of SMARCC1 and its potential as putative therapeutic target in HCC. Methods The mRNA expression profiles and prognostic value of SMARCC1 were analyzed in the Oncomine, UALCAN and Kaplan–Meier Plotter databases. The expression of SMARCC1 and associated clinicopathological factors were further evaluated using a tissue microarray. Differentially expressed genes associated with SMARCC1 in HCC were obtained and analyzed via the LinkedOmics and GEPIA databases and Cytoscape software. To verify the important role of SMARCC1 in HCC, we knocked down and overexpressed SMARCC1 in different hepatic cell lines and conducted several functional experiments. Then, we evaluated the mutation profiles and transcriptional regulators of SMARCC1 using the cBioPortal, COSMIC, CistromeDB and TCGA databases. Finally, we addressed the relationship of SMARCC1 expression with immune cell infiltration via TIMER database analysis. Results Through data mining and tissue microarray verification, we found that the protein and mRNA levels of SMARCC1 are high in tumor tissues, which has remarkable diagnostic value in HCC patients. SMARCC1 and its hub genes showed prognostic value in HCC. Furthermore, we confirmed that SMARCC1 influenced the proliferation, migration, and invasion of HCC cells. Moreover, correlation analyses revealed that SMARCC1 expression was positively correlated with ZBTB40 transcription factors and negatively correlated with the DNA methylation level. Overall, we found that SMARCC1 affects immune infiltration and plays a tumor-promoting role in HCC. Conclusions SMARCC1 is overexpressed and is a putative prognostic predictor in HCC. Due to the tumor-promoting role of SMARCC1, treatments inhibiting DNA methyltransferases and transcription factors or weakening the role of SMARCC1 in immune infiltration might improve the survival of HCC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02413-w.
Collapse
|