1
|
Zhang Y, Lu Y, Yu M, Wang J, Du X, Zhao D, Pian H, He Z, Wu G, Li S, Wang S, Yu D. Transcriptome Profiling Identifies Differentially Expressed Genes in Skeletal Muscle Development in Native Chinese Ducks. Genes (Basel) 2023; 15:52. [PMID: 38254942 PMCID: PMC10815232 DOI: 10.3390/genes15010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
China boasts a rich diversity of indigenous duck species, some of which exhibit desirable economic traits. Here, we generated transcriptome sequencing datasets of breast muscle tissue samples from 1D of four groups: Pekin duck pure breeding group (P), Jinling White duck breeding group (J), P ♂ × J ♀ orthogonal group (PJ) and J ♂ × P ♀ reciprocal-cross group (JP) (n = 3), chosen based on the distinctive characteristics of duck muscle development during the embryonic period. We identified 5053 differentially expressed genes (DEGs) among the four groups. Network prediction analysis showed that ribosome and oxidative phosphorylation-related genes were the most enriched, and muscular protein-related genes were found in the 14-day-old embryonic group. We found that previously characterized functional genes, such as FN1, AGRN, ADNAMST3, APOB and FGF9, were potentially involved in muscle development in 14-day-old embryos. Functional enrichment analysis suggested that genes that participated in molecular function and cell component and key signaling pathways (e.g., hippo, ribosome, oxidative phosphorylation) were significantly enriched in the development of skeletal muscle at 14 days of embryonic age. These results indicate a possible role of muscle metabolism and myoglobin synthesis in skeletal muscle development in both duck parents and hybrids.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Yinglin Lu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Minli Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Jin Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Xubin Du
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Dong Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
- School of Animal Medical, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Huifang Pian
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Zongliang He
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Guansuo Wu
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Shiwei Li
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Sike Wang
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Debing Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| |
Collapse
|
2
|
Hollander JM, Goraltchouk A, Rawal M, Liu J, Luppino F, Zeng L, Seregin A. Adeno-Associated Virus-Delivered Fibroblast Growth Factor 18 Gene Therapy Promotes Cartilage Anabolism. Cartilage 2023; 14:492-505. [PMID: 36879540 PMCID: PMC10807742 DOI: 10.1177/19476035231158774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/21/2023] [Accepted: 01/31/2023] [Indexed: 03/08/2023] Open
Abstract
OBJECTIVE To determine the characterization of chondrogenic properties of adeno-associated virus type 2 (AAV2)-delivered hFGF18, via analysis of effects on primary human chondrocyte proliferation, gene expression, and in vivo cartilage thickness changes in the tibia and meniscus. DESIGN Chondrogenic properties of AAV2-FGF18 were compared with recombinant human FGF18 (rhFGF18) in vitro relative to phosphate-buffered saline (PBS) and AAV2-GFP negative controls. Transcriptome analysis was performed using RNA-seq on primary human chondrocytes treated with rhFGF18 and AAV2-FGF18, relative to PBS. Durability of gene expression was assessed using AAV2-nLuc and in vivo imaging. Chondrogenesis was evaluated by measuring weight-normalized thickness in the tibial plateau and the white zone of the anterior horn of the medial meniscus in Sprague-Dawley rats. RESULTS AAV2-FGF18 elicits chondrogenesis by promoting proliferation and upregulation of hyaline cartilage-associated genes, including COL2A1 and HAS2, while downregulating fibrocartilage-associated COL1A1. This activity translates to statistically significant, dose-dependent increases in cartilage thickness in vivo within the area of the tibial plateau, following a single intra-articular injection of the AAV2-FGF18 or a regimen of 6 twice-weekly injections of rhFGF18 protein relative to AAV2-GFP. In addition, we observed AAV2-FGF18-induced and rhFGF18-induced increases in cartilage thickness of the anterior horn of the medial meniscus. Finally, the single-injection AAV2-delivered hFGF18 offers a potential safety advantage over the multi-injection protein treatment as evidenced by reduced joint swelling over the study period. CONCLUSION AAV2-delivered hFGF18 represents a promising strategy for the restoration of hyaline cartilage by promoting extracellular matrix production, chondrocyte proliferation, and increasing articular and meniscal cartilage thickness in vivo after a single intra-articular injection.
Collapse
Affiliation(s)
- Judith M. Hollander
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | | | - Miraj Rawal
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Jingshu Liu
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | | | - Li Zeng
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
3
|
Glycine-Serine-Threonine Metabolic Axis Delays Intervertebral Disc Degeneration through Antioxidant Effects: An Imaging and Metabonomics Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5579736. [PMID: 34484565 PMCID: PMC8416401 DOI: 10.1155/2021/5579736] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022]
Abstract
Although intervertebral disc degeneration (IDD) can be described as different stages of change through biological methods, this long and complex process cannot be defined in stages by single or simple combination of biological techniques. Under the background of the development of nuclear magnetic resonance (NMR) technology and the emerging metabonomics, we based on animal models and expanded to the study of clinical human degeneration models. The characteristics of different stages of IDD were analyzed by omics. Omics imaging combined with histology, cytology, and proteomics was used for screening of the intervertebral disc (IVD) of research subjects. Furthermore, mass spectrometry nontargeted metabolomics was used to explore profile of metabolites at different stages of the IDD process, to determine differential metabolic pathways and metabolites. NMR spectroscopy was used to qualitatively and quantitatively identify markers of degeneration. NMR was combined with mass spectrometry metabolomics to explore metabolic pathways. Metabolic pathways were determined through protein molecular biology and histocytology of the different groups. Distinguishing advantages of magnetic resonance spectroscopy (MRS) for analysis of metabolites and effective reflection of structural integrity and water molecule metabolism through diffusion tensor imaging (DTI) were further used to verify the macrometabolism profile during degeneration. A corresponding model of in vitro metabolomics and in vivo omics imaging was established. The findings of this study show that a series of metabolic pathways associated with the glycine-serine-threonine (Gly-Ser-Thr) metabolic axis affects carbohydrate patterns and energy utilization efficiency and ultimately delays disc degeneration through antioxidant effects.
Collapse
|
4
|
Théret N, Bouezzeddine F, Azar F, Diab-Assaf M, Legagneux V. ADAM and ADAMTS Proteins, New Players in the Regulation of Hepatocellular Carcinoma Microenvironment. Cancers (Basel) 2021; 13:cancers13071563. [PMID: 33805340 PMCID: PMC8037375 DOI: 10.3390/cancers13071563] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Members of the adamalysin family are multi-domain proteins involved in many cancer-related functions. In this review, we will examine the literature on the involvement of adamalysins in hepatocellular carcinoma progression and their importance in the tumor microenvironment where they regulate the inflammatory response and the epithelial–mesenchymal transition. We complete this review with an analysis of adamalysin expression in a large cohort of patients with hepatocellular carcinoma from The Cancer Genome Atlas (TCGA) database. These original results give a new insight into the involvement of all adamalysins in the primary liver cancer. Abstract The tumor microenvironment plays a major role in tumor growth, invasion and resistance to chemotherapy, however understanding how all actors from microenvironment interact together remains a complex issue. The tumor microenvironment is classically represented as three closely connected components including the stromal cells such as immune cells, fibroblasts, adipocytes and endothelial cells, the extracellular matrix (ECM) and the cytokine/growth factors. Within this space, proteins of the adamalysin family (ADAM for a disintegrin and metalloproteinase; ADAMTS for ADAM with thrombospondin motifs; ADAMTSL for ADAMTS-like) play critical roles by modulating cell–cell and cell–ECM communication. During last decade, the implication of adamalysins in the development of hepatocellular carcinoma (HCC) has been supported by numerous studies however the functional characterization of most of them remain unsettled. In the present review we propose both an overview of the literature and a meta-analysis of adamalysins expression in HCC using data generated by The Cancer Genome Atlas (TCGA) Research Network.
Collapse
Affiliation(s)
- Nathalie Théret
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
- Correspondence:
| | - Fidaa Bouezzeddine
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Fida Azar
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| | - Mona Diab-Assaf
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| |
Collapse
|
5
|
Koshiba S, Motoike IN, Saigusa D, Inoue J, Aoki Y, Tadaka S, Shirota M, Katsuoka F, Tamiya G, Minegishi N, Fuse N, Kinoshita K, Yamamoto M. Identification of critical genetic variants associated with metabolic phenotypes of the Japanese population. Commun Biol 2020; 3:662. [PMID: 33177615 PMCID: PMC7659008 DOI: 10.1038/s42003-020-01383-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
We performed a metabolome genome-wide association study for the Japanese population in the prospective cohort study of Tohoku Medical Megabank. By combining whole-genome sequencing and nontarget metabolome analyses, we identified a large number of novel associations between genetic variants and plasma metabolites. Of the identified metabolite-associated genes, approximately half have already been shown to be involved in various diseases. We identified metabolite-associated genes involved in the metabolism of xenobiotics, some of which are from intestinal microorganisms, indicating that the identified genetic variants also markedly influence the interaction between the host and symbiotic bacteria. We also identified five associations that appeared to be female-specific. A number of rare variants that influence metabolite levels were also found, and combinations of common and rare variants influenced the metabolite levels more profoundly. These results support our contention that metabolic phenotyping provides important insights into how genetic and environmental factors provoke human diseases.
Collapse
Affiliation(s)
- Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| | - Ikuko N Motoike
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09, Aramaki Aza-Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Jin Inoue
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yuichi Aoki
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09, Aramaki Aza-Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Shu Tadaka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09, Aramaki Aza-Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Matsuyuki Shirota
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Fumiki Katsuoka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Naoko Minegishi
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Nobuo Fuse
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09, Aramaki Aza-Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
- Graduate School of Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| |
Collapse
|
6
|
Nandadasa S, Szafron JM, Pathak V, Murtada SI, Kraft CM, O'Donnell A, Norvik C, Hughes C, Caterson B, Domowicz MS, Schwartz NB, Tran-Lundmark K, Veigl M, Sedwick D, Philipson EH, Humphrey JD, Apte SS. Vascular dimorphism ensured by regulated proteoglycan dynamics favors rapid umbilical artery closure at birth. eLife 2020; 9:e60683. [PMID: 32909945 PMCID: PMC7529456 DOI: 10.7554/elife.60683] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/09/2020] [Indexed: 01/29/2023] Open
Abstract
The umbilical artery lumen closes rapidly at birth, preventing neonatal blood loss, whereas the umbilical vein remains patent longer. Here, analysis of umbilical cords from humans and other mammals identified differential arterial-venous proteoglycan dynamics as a determinant of these contrasting vascular responses. The umbilical artery, but not the vein, has an inner layer enriched in the hydrated proteoglycan aggrecan, external to which lie contraction-primed smooth muscle cells (SMC). At birth, SMC contraction drives inner layer buckling and centripetal displacement to occlude the arterial lumen, a mechanism revealed by biomechanical observations and confirmed by computational analyses. This vascular dimorphism arises from spatially regulated proteoglycan expression and breakdown. Mice lacking aggrecan or the metalloprotease ADAMTS1, which degrades proteoglycans, demonstrate their opposing roles in umbilical vascular dimorphism, including effects on SMC differentiation. Umbilical vessel dimorphism is conserved in mammals, suggesting that differential proteoglycan dynamics and inner layer buckling were positively selected during evolution.
Collapse
Affiliation(s)
- Sumeda Nandadasa
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Jason M Szafron
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | - Vai Pathak
- Case Comprehensive Cancer Center, Case Western Reserve UniversityClevelandUnited States
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | - Caroline M Kraft
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Anna O'Donnell
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Christian Norvik
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund UniversityLundSweden
| | - Clare Hughes
- The Sir Martin Evans Building, School of Biosciences, Cardiff UniversityCardiffUnited Kingdom
| | - Bruce Caterson
- The Sir Martin Evans Building, School of Biosciences, Cardiff UniversityCardiffUnited Kingdom
| | | | - Nancy B Schwartz
- Department of Pediatrics, University of ChicagoChicagoUnited States
| | - Karin Tran-Lundmark
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund UniversityLundSweden
| | - Martina Veigl
- Case Comprehensive Cancer Center, Case Western Reserve UniversityClevelandUnited States
- Department of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - David Sedwick
- Department of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Elliot H Philipson
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
- The Women's Health Institute, Department of Obstetrics and Gynecology, Cleveland ClinicClevelandUnited States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| |
Collapse
|
7
|
Holdener BC, Percival CJ, Grady RC, Cameron DC, Berardinelli SJ, Zhang A, Neupane S, Takeuchi M, Jimenez-Vega JC, Uddin SMZ, Komatsu DE, Honkanen R, Dubail J, Apte SS, Sato T, Narimatsu H, McClain SA, Haltiwanger RS. ADAMTS9 and ADAMTS20 are differentially affected by loss of B3GLCT in mouse model of Peters plus syndrome. Hum Mol Genet 2020; 28:4053-4066. [PMID: 31600785 DOI: 10.1093/hmg/ddz225] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 01/15/2023] Open
Abstract
Peters plus syndrome (MIM #261540 PTRPLS), characterized by defects in eye development, prominent forehead, hypertelorism, short stature and brachydactyly, is caused by mutations in the β3-glucosyltransferase (B3GLCT) gene. Protein O-fucosyltransferase 2 (POFUT2) and B3GLCT work sequentially to add an O-linked glucose β1-3fucose disaccharide to properly folded thrombospondin type 1 repeats (TSRs). Forty-nine proteins are predicted to be modified by POFUT2, and nearly half are members of the ADAMTS superfamily. Previous studies suggested that O-linked fucose is essential for folding and secretion of POFUT2-modified proteins and that B3GLCT-mediated extension to the disaccharide is essential for only a subset of targets. To test this hypothesis and gain insight into the origin of PTRPLS developmental defects, we developed and characterized two mouse B3glct knockout alleles. Using these models, we tested the role of B3GLCT in enabling function of ADAMTS9 and ADAMTS20, two highly conserved targets whose functions are well characterized in mouse development. The mouse B3glct mutants developed craniofacial and skeletal abnormalities comparable to PTRPLS. In addition, we observed highly penetrant hydrocephalus, white spotting and soft tissue syndactyly. We provide strong genetic and biochemical evidence that hydrocephalus and white spotting in B3glct mutants resulted from loss of ADAMTS20, eye abnormalities from partial reduction of ADAMTS9 and cleft palate from loss of ADAMTS20 and partially reduced ADAMTS9 function. Combined, these results provide compelling evidence that ADAMTS9 and ADAMTS20 were differentially sensitive to B3GLCT inactivation and suggest that the developmental defects in PTRPLS result from disruption of a subset of highly sensitive POFUT2/B3GLCT targets such as ADAMTS20.
Collapse
Affiliation(s)
- Bernadette C Holdener
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | | | - Richard C Grady
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel C Cameron
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Steven J Berardinelli
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Ao Zhang
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Sanjiv Neupane
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Megumi Takeuchi
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | - Sardar M Z Uddin
- Department of Orthopaedics, Stony Brook University, Stony Brook, NY 11794, USA
| | - David E Komatsu
- Department of Orthopaedics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Robert Honkanen
- Department of Ophthalmology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Johanne Dubail
- Department of Biomedical Engineering, Cleveland Clinic Lerner Institute, Cleveland, OH 44195, USA
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Institute, Cleveland, OH 44195, USA
| | - Takashi Sato
- National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Hisashi Narimatsu
- National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Steve A McClain
- Department of Dermatology and Department of Emergency Medicine, Stony Brook University, Stony Brook, NY 11794, USA.,Department of Emergency Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
8
|
Jiménez-Meléndez A, Ramakrishnan C, Hehl AB, Russo G, Álvarez-García G. RNA-Seq Analyses Reveal That Endothelial Activation and Fibrosis Are Induced Early and Progressively by Besnoitia besnoiti Host Cell Invasion and Proliferation. Front Cell Infect Microbiol 2020; 10:218. [PMID: 32500038 PMCID: PMC7242738 DOI: 10.3389/fcimb.2020.00218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of bovine besnoitiosis and the molecular bases that govern disease progression remain to be elucidated. Thus, we have employed an in vitro model of infection based on primary bovine aortic endothelial cells (BAEC), target cells during the acute infection. Host-parasite interactions were investigated by RNA-Seq at two post-infection (pi) time points: 12 hpi, when tachyzoites have already invaded host cells, and 32 hpi, when tachyzoites have replicated for at least two generations. Additionally, the gene expression profile of B. besnoiti tachyzoites was studied at both pi time points. Up to 446 differentially expressed B. taurus genes (DEGs) were found in BAEC between both pi time points: 249 DEGs were up-regulated and 197 DEGs were down-regulated at 32 hpi. Upregulation of different genes encoding cytokines, chemokines, leukocyte adhesion molecules predominantly at 12 hpi implies an activation of endothelial cells, whilst upregulation of genes involved in angiogenesis and extracellular matrix organization was detected at both time points. NF-κB and TNF-α signaling pathways appeared to be mainly modulated upon infection, coordinating the expression of several effector proteins with proinflammatory and pro-fibrotic phenotypes. These mediators are thought to be responsible for macrophage recruitment setting the basis for chronic inflammation and fibrosis characteristic of chronic besnoitiosis. Angiogenesis regulation also predominated, and this multistep process was evidenced by the upregulation of markers involved in both early (e.g., growth factors and matrix metalloproteinases) and late steps (e.g., integrins and vasohibin). Besnoitia besnoiti ortholog genes present in other Toxoplasmatinae members and involved in the lytic cycle have shown to be differentially expressed among the two time points studied, with a higher expression at 32 hpi (e.g., ROP40, ROP5B, MIC1, MIC10). This study gives molecular clues on B. besnoiti- BAECs interaction and shows the progression of type II endothelial cell activation upon parasite invasion and proliferation.
Collapse
Affiliation(s)
- Alejandro Jiménez-Meléndez
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | | | - Adrian B Hehl
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | | | - Gema Álvarez-García
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
9
|
ADAMTS-15 Has a Tumor Suppressor Role in Prostate Cancer. Biomolecules 2020; 10:biom10050682. [PMID: 32354091 PMCID: PMC7277637 DOI: 10.3390/biom10050682] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/18/2022] Open
Abstract
Extracellular matrix remodeling has emerged as an important factor in many cancers. Proteoglycans, including versican (VCAN), are regulated via cleavage by the proteolytic actions of A Disintegrin-like And Metalloproteinase domain with Thrombospondin-1 motif (ADAMTS) family members. Alterations in the balance between Proteoglycans and ADAMTS enzymes have been proposed to contribute to cancer progression. Here, we analyzed the expression of ADAMTS-15 in human prostate cancer, and investigated the effects of enforced expression in prostate cancer cell lines. ADAMTS-15 was found to be expressed in human prostate cancer biopsies with evidence of co-localization with VCAN and its bioactive cleavage fragment versikine. Enforced expression of ADAMTS-15, but not a catalytically-inactive version, decreased cell proliferation and migration of the ‘castrate-resistant’ PC3 prostate cancer cell line in vitro, with survival increased. Analysis of ‘androgen-responsive’ LNCaP prostate cancer cells in vivo in NOD/SCID mice revealed that ADAMTS-15 expression caused slower growing tumors, which resulted in increased survival. This was not observed in castrated mice or with cells expressing catalytically-inactive ADAMTS-15. Collectively, this research identifies the enzymatic function of ADAMTS-15 as having a tumor suppressor role in prostate cancer, possibly in concert with androgens, and that VCAN represents a likely key substrate, highlighting potential new options for the clinic.
Collapse
|
10
|
Mohamedi Y, Fontanil T, Cobo T, Cal S, Obaya AJ. New Insights into ADAMTS Metalloproteases in the Central Nervous System. Biomolecules 2020; 10:biom10030403. [PMID: 32150898 PMCID: PMC7175268 DOI: 10.3390/biom10030403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Components of the extracellular matrix (ECM) are key players in regulating cellular functions throughout the whole organism. In fact, ECM components not only participate in tissue organization but also contribute to processes such as cellular maintenance, proliferation, and migration, as well as to support for various signaling pathways. In the central nervous system (CNS), proteoglycans of the lectican family, such as versican, aggrecan, brevican, and neurocan, are important constituents of the ECM. In recent years, members of this family have been found to be involved in the maintenance of CNS homeostasis and to participate directly in processes such as the organization of perineural nets, the regulation of brain plasticity, CNS development, brain injury repair, axonal guidance, and even the altering of synaptic responses. ADAMTSs are a family of “A disintegrin and metalloproteinase with thrombospondin motifs” proteins that have been found to be involved in a multitude of processes through the degradation of lecticans and other proteoglycans. Recently, alterations in ADAMTS expression and activity have been found to be involved in neuronal disorders such as stroke, neurodegeneration, schizophrenia, and even Alzheimer’s disease, which in turn may suggest their potential use as therapeutic targets. Herein, we summarize the different roles of ADAMTSs in regulating CNS events through interactions and the degradation of ECM components (more specifically, the lectican family of proteoglycans).
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Departamento de Investigación, Instituto Ordóñez, 33012 Oviedo, Asturias, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain;
- Instituto Asturiano de Odontología, 33006 Oviedo, Asturias, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Alvaro J. Obaya
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Correspondence:
| |
Collapse
|
11
|
Paiva KBS, Maas CS, dos Santos PM, Granjeiro JM, Letra A. Extracellular Matrix Composition and Remodeling: Current Perspectives on Secondary Palate Formation, Cleft Lip/Palate, and Palatal Reconstruction. Front Cell Dev Biol 2019; 7:340. [PMID: 31921852 PMCID: PMC6923686 DOI: 10.3389/fcell.2019.00340] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Craniofacial development comprises a complex process in humans in which failures or disturbances frequently lead to congenital anomalies. Cleft lip with/without palate (CL/P) is a common congenital anomaly that occurs due to variations in craniofacial development genes, and may occur as part of a syndrome, or more commonly in isolated forms (non-syndromic). The etiology of CL/P is multifactorial with genes, environmental factors, and their potential interactions contributing to the condition. Rehabilitation of CL/P patients requires a multidisciplinary team to perform the multiple surgical, dental, and psychological interventions required throughout the patient's life. Despite progress, lip/palatal reconstruction is still a major treatment challenge. Genetic mutations and polymorphisms in several genes, including extracellular matrix (ECM) genes, soluble factors, and enzymes responsible for ECM remodeling (e.g., metalloproteinases), have been suggested to play a role in the etiology of CL/P; hence, these may be considered likely targets for the development of new preventive and/or therapeutic strategies. In this context, investigations are being conducted on new therapeutic approaches based on tissue bioengineering, associating stem cells with biomaterials, signaling molecules, and innovative technologies. In this review, we discuss the role of genes involved in ECM composition and remodeling during secondary palate formation and pathogenesis and genetic etiology of CL/P. We also discuss potential therapeutic approaches using bioactive molecules and principles of tissue bioengineering for state-of-the-art CL/P repair and palatal reconstruction.
Collapse
Affiliation(s)
- Katiúcia Batista Silva Paiva
- Laboratory of Extracellular Matrix Biology and Cellular Interaction, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Clara Soeiro Maas
- Laboratory of Extracellular Matrix Biology and Cellular Interaction, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Pâmella Monique dos Santos
- Laboratory of Extracellular Matrix Biology and Cellular Interaction, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José Mauro Granjeiro
- Clinical Research Laboratory in Dentistry, Federal Fluminense University, Niterói, Brazil
- Directory of Life Sciences Applied Metrology, National Institute of Metrology, Quality and Technology, Duque de Caxias, Brazil
| | - Ariadne Letra
- Center for Craniofacial Research, UTHealth School of Dentistry at Houston, Houston, TX, United States
- Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX, United States
- Department of Diagnostic and Biomedical Sciences, UTHealth School of Dentistry at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Mendoza-Topaz C, Nelson G, Howard G, Hafner S, Rademacher P, Frick M, Nichols BJ. Cells respond to deletion of CAV1 by increasing synthesis of extracellular matrix. PLoS One 2018; 13:e0205306. [PMID: 30346954 PMCID: PMC6197626 DOI: 10.1371/journal.pone.0205306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
A range of cellular functions have been attributed to caveolae, flask-like invaginations of the plasma membrane. Here, we have used RNA-seq to achieve quantitative transcriptional profiling of primary embryonic fibroblasts from caveolin 1 knockout mice (CAV1-/- MEFs), and thereby to gain hypothesis-free insight into how these cells respond to the absence of caveolae. Components of the extracellular matrix were decisively over-represented within the set of genes displaying altered expression in CAV1-/- MEFs when compared to congenic wild-type controls. This was confirmed biochemically and by imaging for selected examples. Up-regulation of components of the extracellular matrix was also observed in a second cell line, NIH-3T3 cells genome edited to delete CAV1. Up-regulation of components of the extracellular matrix was detected in vivo by assessing collagen deposition and compliance of CAV1-/- lungs. We discuss the implications of these findings in terms of the cellular function of caveolae.
Collapse
Affiliation(s)
- C. Mendoza-Topaz
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - G. Nelson
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - G. Howard
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - S. Hafner
- Institute of Pathophysiological Anesthesiology and Process Engineering, University of Ulm, Ulm, Germany
| | - P. Rademacher
- Institute of Pathophysiological Anesthesiology and Process Engineering, University of Ulm, Ulm, Germany
| | - M. Frick
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - B. J. Nichols
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Dancevic CM, Gibert Y, Berger J, Smith AD, Liongue C, Stupka N, Ward AC, McCulloch DR. The ADAMTS5 Metzincin Regulates Zebrafish Somite Differentiation. Int J Mol Sci 2018. [PMID: 29518972 PMCID: PMC5877627 DOI: 10.3390/ijms19030766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The ADAMTS5 metzincin, a secreted zinc-dependent metalloproteinase, modulates the extracellular matrix (ECM) during limb morphogenesis and other developmental processes. Here, the role of ADAMTS5 was investigated by knockdown of zebrafish adamts5 during embryogenesis. This revealed impaired Sonic Hedgehog (Shh) signaling during somite patterning and early myogenesis. Notably, synergistic regulation of myod expression by ADAMTS5 and Shh during somite differentiation was observed. These roles were not dependent upon the catalytic activity of ADAMTS5. These data identify a non-enzymatic function for ADAMTS5 in regulating an important cell signaling pathway that impacts on muscle development, with implications for musculoskeletal diseases in which ADAMTS5 and Shh have been associated.
Collapse
Affiliation(s)
- Carolyn M Dancevic
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Yann Gibert
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia.
| | - Adam D Smith
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Clifford Liongue
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Nicole Stupka
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Alister C Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| | - Daniel R McCulloch
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia.
- Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| |
Collapse
|