1
|
Sun R, Duan D, Li R. Transcriptome Sequencing Identifies Abnormal lncRNAs and mRNAs and Reveals Potentially Hub Immune-Related mRNA in Osteoporosis with Vertebral Fracture. Clin Interv Aging 2024; 19:203-217. [PMID: 38352274 PMCID: PMC10863500 DOI: 10.2147/cia.s441251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
Background Recent studies have put forward the viewpoint of "bone immunology", which holds that the immune system and immune factors play an important regulatory role in the occurrence and development of osteoporosis. This study was intended to identify genetic characteristics of differentially expressed immune-related mRNA and lncRNA in patients combined with osteoporosis and vertebral fracture. Methods The peripheral blood samples were obtained from 3 groups of subjects: healthy control (HC), osteoporosis patients without vertebral fracture (OWF), and osteoporosis patients combined with vertebral fracture (OVF). The data were integrated to obtain differentially expressed mRNAs (DEmRNAs) and differentially expressed lncRNAs (DElncRNAs). Subsequently, the protein-protein interaction (PPI) networks were constructed. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analyses were performed. Cytoscape-cytoHubba plug-in was used to identify key DEmRNAs. Furthermore, lncRNA-miRNA-mRNA, mRNA-lncRNA co-expression and transcription factors (TFs) networks were constructed. In addition, real-time PCR verification was performed. Results Totally of 3378 lncRNA-mRNA pairs were obtained, and the lncRNA co-expressed mRNA was mainly enriched in immune-related pathways, especially in GO-biological process (GO-BP) analysis. A total of 8 hub immune-related DEmRNAs were obtained, including IL18R1, IL18RAP, SLC11A1, CSF2RA, CCR3, IL1R2, PGLYRP1, and IL1R1. The TFs network showed that 8 hub immune-related DEmRNAs had interacting TFs. The co-expression network showed that 7 hub immune-related DEmRNAs (IL18R1, IL18RAP, SLC11A1, CSF2RA, IL-1R2, PGLYRP1, and IL1R1) had lncRNA-mRNA co-expression relationship. In addition, the lncRNA-miRNA-mRNA network includes 32 miRNAs, 7 hub immune-related mRNAs (IL18R1, IL18RAP, CSF2RA, CCR3, IL1R2, PGLYRP1, and IL1R1), and 11 lncRNAs. Conclusion Our study provides a novel and in-depth identification of co-expressed mRNAs and lncRNAs in patients combined with osteoporosis and vertebral fracture at a molecular level. This may provide new candidate biomarkers for the diagnosis of patients with high-risk fractures in the future.
Collapse
Affiliation(s)
- Rongxin Sun
- Department of Orthopaedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Province, People’s Republic of China
| | - Desheng Duan
- Second Department of Orthopaedics, Third People’s Hospital of Anyang City, Anyang City, Henan Province, People’s Republic of China
| | - Renzeng Li
- Second Department of Orthopaedics, Third People’s Hospital of Anyang City, Anyang City, Henan Province, People’s Republic of China
| |
Collapse
|
2
|
Goel RK, Kim N, Lukong KE. Seeking a better understanding of the non-receptor tyrosine kinase, SRMS. Heliyon 2023; 9:e16421. [PMID: 37251450 PMCID: PMC10220380 DOI: 10.1016/j.heliyon.2023.e16421] [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: 01/15/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023] Open
Abstract
SRMS (Src-Related kinase lacking C-terminal regulatory tyrosine and N-terminal Myristoylation Sites) is a non-receptor tyrosine kinase first reported in a 1994 screen for genes regulating murine neural precursor cells. SRMS, pronounced "Shrims", lacks the C-terminal regulatory tyrosine critical for the regulation of the enzymatic activity of Src-family kinases (SFKs). Another remarkable characteristic of SRMS is its localization into distinct SRMS cytoplasmic punctae (SCPs) or GREL (Goel Raghuveera-Erique Lukong) bodies, a pattern not observed in the SFKs. This unique subcellular localization of SRMS could dictate its cellular targets, proteome, and potentially, substrates. However, the function of SRMS is still relatively unknown. Further, how is its activity regulated and by what cellular targets? Studies have emerged highlighting the potential role of SRMS in autophagy and in regulating the activation of BRK/PTK6. Potential novel cellular substrates have also been identified, including DOK1, vimentin, Sam68, FBKP51, and OTUB1. Recent studies have also demonstrated the potential role of the kinase in various cancers, including gastric and colorectal cancers and platinum resistance in ovarian cancer. This review discusses the advancements made in SRMS-related biology to date and the path to understanding the cellular and physiological significance of the kinase.
Collapse
Affiliation(s)
- Raghuveera Kumar Goel
- Center for Network Systems Biology, Boston University, Boston, MA, USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Nayoung Kim
- Department of Biochemistry, Microbiology, and Immunology, 107 Wiggins Road, Health Sciences Building, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology, and Immunology, 107 Wiggins Road, Health Sciences Building, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| |
Collapse
|
3
|
Hunt BG, Fox LH, Davis JC, Jones A, Lu Z, Waltz SE. An Introduction and Overview of RON Receptor Tyrosine Kinase Signaling. Genes (Basel) 2023; 14:517. [PMID: 36833444 PMCID: PMC9956929 DOI: 10.3390/genes14020517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
RON is a receptor tyrosine kinase (RTK) of the MET receptor family that is canonically involved in mediating growth and inflammatory signaling. RON is expressed at low levels in a variety of tissues, but its overexpression and activation have been associated with malignancies in multiple tissue types and worse patient outcomes. RON and its ligand HGFL demonstrate cross-talk with other growth receptors and, consequentially, positions RON at the intersection of numerous tumorigenic signaling programs. For this reason, RON is an attractive therapeutic target in cancer research. A better understanding of homeostatic and oncogenic RON activity serves to enhance clinical insights in treating RON-expressing cancers.
Collapse
Affiliation(s)
- Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Levi H. Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - James C. Davis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Angelle Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Zhixin Lu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
- Research Service, Cincinnati Veterans Affairs Hospital Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
4
|
Erythropoietin Activates Autophagy to Regulate Apoptosis and Angiogenesis of Periodontal Ligament Stem Cells via the Akt/ERK1/2/BAD Signaling Pathway under Inflammatory Microenvironment. Stem Cells Int 2022; 2022:9806887. [PMID: 36199627 PMCID: PMC9527112 DOI: 10.1155/2022/9806887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/18/2022] [Accepted: 09/02/2022] [Indexed: 11/18/2022] Open
Abstract
Background. Angiogenic tissue engineering is a vital problem waiting to be settled for periodontal regeneration. Erythropoietin, a multieffect cytokine, has been reported as a protective factor for cell fate. According to our previous study, erythropoietin has a significantly angiogenic effect on periodontal ligament stem cells. To further explore its potential effects and mechanism, we studied biological behaviors of periodontal ligament stem cells under inflammatory microenvironment induced by different concentrations (0, 10, 20, 50, and 100 ng/mL) of tumor necrosis factor-α (TNF-α) and examined how different concentrations (0, 5, 10, 20, and 50 IU/mL) of erythropoietin changed biological behaviors of periodontal ligament stem cells. Materials and Methods. Cell Counting Kit-8 was used for cell proliferation assay. Annexin V-PI-FITC was used for cell apoptosis through flow cytometry. Matrigel plug was adopted to measure the angiogenic capacity in vitro. RNA sequencing was used to detect the downstream signaling pathway. Quantitative real-time polymerase chain reaction was conducted to examine mRNA expression level. Western blot and immunofluorescence were applied to testify the protein expression level. Results. Periodontal ligament stem cells upregulated apoptosis and suppressed autophagy and angiogenesis under inflammatory microenvironment. Erythropoietin could activate autophagy to rescue apoptosis and angiogenesis levels of periodontal ligament stem cells through the Akt/Erk1/2/BAD signaling pathway under inflammatory microenvironment. Conclusions. Erythropoietin could protect periodontal ligament stem cells from inflammatory microenvironment, which provided a novel theory for periodontal regeneration.
Collapse
|
5
|
Hung CH, Wang KY, Liou YH, Wang JP, Huang AYS, Lee TL, Jiang ST, Liao NS, Shyu YC, Shen CKJ. Negative Regulation of the Differentiation of Flk2 - CD34 - LSK Hematopoietic Stem Cells by EKLF/KLF1. Int J Mol Sci 2020; 21:E8448. [PMID: 33182781 PMCID: PMC7697791 DOI: 10.3390/ijms21228448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Erythroid Krüppel-like factor (EKLF/KLF1) was identified initially as a critical erythroid-specific transcription factor and was later found to be also expressed in other types of hematopoietic cells, including megakaryocytes and several progenitors. In this study, we have examined the regulatory effects of EKLF on hematopoiesis by comparative analysis of E14.5 fetal livers from wild-type and Eklf gene knockout (KO) mouse embryos. Depletion of EKLF expression greatly changes the populations of different types of hematopoietic cells, including, unexpectedly, the long-term hematopoietic stem cells Flk2- CD34- Lin- Sca1+ c-Kit+ (LSK)-HSC. In an interesting correlation, Eklf is expressed at a relatively high level in multipotent progenitor (MPP). Furthermore, EKLF appears to repress the expression of the colony-stimulating factor 2 receptor β subunit (CSF2RB). As a result, Flk2- CD34- LSK-HSC gains increased differentiation capability upon depletion of EKLF, as demonstrated by the methylcellulose colony formation assay and by serial transplantation experiments in vivo. Together, these data demonstrate the regulation of hematopoiesis in vertebrates by EKLF through its negative regulatory effects on the differentiation of the hematopoietic stem and progenitor cells, including Flk2- CD34- LSK-HSCs.
Collapse
Affiliation(s)
- Chun-Hao Hung
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Keh-Yang Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Jing-Ping Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Anna Yu-Szu Huang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Tung-Liang Lee
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Si-Tse Jiang
- Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Tainan 74147, Taiwan;
| | - Nah-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yu-Chiau Shyu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City 333, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- The PhD Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 115, Taiwan
| |
Collapse
|
6
|
Fan T, Qu R, Yu Q, Sun B, Jiang X, Yang Y, Huang X, Zhou Z, Ouyang J, Zhong S, Dai J. Bioinformatics analysis of the biological changes involved in the osteogenic differentiation of human mesenchymal stem cells. J Cell Mol Med 2020; 24:7968-7978. [PMID: 32463168 PMCID: PMC7348183 DOI: 10.1111/jcmm.15429] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 12/17/2022] Open
Abstract
The mechanisms underlying the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) remain unclear. In the present study, we aimed to identify the key biological processes during osteogenic differentiation. To this end, we downloaded three microarray data sets from the Gene Expression Omnibus (GEO) database: GSE12266, GSE18043 and GSE37558. Differentially expressed genes (DEGs) were screened using the limma package, and enrichment analysis was performed. Protein-protein interaction network (PPI) analysis and visualization analysis were performed with STRING and Cytoscape. A total of 240 DEGs were identified, including 147 up-regulated genes and 93 down-regulated genes. Functional enrichment and pathways of the present DEGs include extracellular matrix organization, ossification, cell division, spindle and microtubule. Functional enrichment analysis of 10 hub genes showed that these genes are mainly enriched in microtubule-related biological changes, that is sister chromatid segregation, microtubule cytoskeleton organization involved in mitosis, and spindle microtubule. Moreover, immunofluorescence and Western blotting revealed dramatic quantitative and morphological changes in the microtubules during the osteogenic differentiation of human adipose-derived stem cells. In summary, the present results provide novel insights into the microtubule- and cytoskeleton-related biological process changes, identifying candidates for the further study of osteogenic differentiation of the mesenchymal stem cells.
Collapse
Affiliation(s)
- Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qinghe Yu
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Xin Jiang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Zhitao Zhou
- Central Laboratory, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Liu G, Xie Y, Su J, Qin H, Wu H, Li K, Yu B, Zhang X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone. FASEB J 2019; 33:11137-11147. [PMID: 31298955 DOI: 10.1096/fj.201900436rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
So far, there has been no effective cure for osteoporotic cortical bone, the most significant change in long bone structure during aging and the main cause of bone fragility fractures, because its underlying molecular and cellular mechanisms remain largely unknown. We used 3- and 15-mo-old mice as well as 15-mo-old mice treated with vehicle and gefitinib to evaluate structural, cellular, and molecular changes in cortical bone. We found that the senescence of osteoprogenitors was increased, whereas the expression of phosphorylated epidermal growth factor receptor (EGFR) on the endosteal surface of cortical bone down-regulated in middle-aged 15-mo-old mice compared with young 3-mo-old mice. Further decreasing EGFR signaling by gefitinib treatment in middle-aged mice resulted in promoted senescence of osteoprogenitors and accelerated cortical bone degeneration. Moreover, inhibiting EGFR signaling suppressed the expression of enhancer of zeste homolog 2 (Ezh2), the repressor of cell senescence-inducer genes, through ERK1/2 pathway, thereby promoting senescence in osteoprogenitors. Down-regulated EGFR signaling plays a physiologically significant role during aging by reducing Ezh2 expression, leading to the senescence of osteoprogenitors and the decline in bone formation on the endosteal surface of cortical bone.-Liu, G., Xie, Y., Su, J., Qin, H., Wu, H., Li, K., Yu, B., Zhang, X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone.
Collapse
Affiliation(s)
- Guanqiao Liu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongheng Xie
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianwen Su
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanjun Qin
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangtian Wu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaiqun Li
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianrong Zhang
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Venkatesan JK, Moutos FT, Rey-Rico A, Estes BT, Frisch J, Schmitt G, Madry H, Guilak F, Cucchiarini M. Chondrogenic Differentiation Processes in Human Bone-Marrow Aspirates Seeded in Three-Dimensional-Woven Poly(ɛ-Caprolactone) Scaffolds Enhanced by Recombinant Adeno-Associated Virus-Mediated SOX9 Gene Transfer. Hum Gene Ther 2018; 29:1277-1286. [PMID: 29717624 DOI: 10.1089/hum.2017.165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Combining gene therapy approaches with tissue engineering procedures is an active area of translational research for the effective treatment of articular cartilage lesions, especially to target chondrogenic progenitor cells such as those derived from the bone marrow. This study evaluated the effect of genetically modifying concentrated human mesenchymal stem cells from bone marrow to induce chondrogenesis by recombinant adeno-associated virus (rAAV) vector gene transfer of the sex-determining region Y-type high-mobility group box 9 (SOX9) factor upon seeding in three-dimensional-woven poly(ɛ-caprolactone; PCL) scaffolds that provide mechanical properties mimicking those of native articular cartilage. Prolonged, effective SOX9 expression was reported in the constructs for at least 21 days, the longest time point evaluated, leading to enhanced metabolic and chondrogenic activities relative to the control conditions (reporter lacZ gene transfer or absence of vector treatment) but without affecting the proliferative activities in the samples. The application of the rAAV SOX9 vector also prevented undesirable hypertrophic and terminal differentiation in the seeded concentrates. As bone marrow is readily accessible during surgery, such findings reveal the therapeutic potential of providing rAAV-modified marrow concentrates within three-dimensional-woven PCL scaffolds for repair of focal cartilage lesions.
Collapse
Affiliation(s)
- Jagadeesh K Venkatesan
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| | | | - Ana Rey-Rico
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| | | | - Janina Frisch
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| | - Gertrud Schmitt
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| | - Henning Madry
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| | - Farshid Guilak
- 2 Cytex Therapeutics, Inc. , Durham, North Carolina.,3 Departments of Orthopedic Surgery, Developmental Biology, and Biomedical Engineering, Washington University and Shriners Hospitals for Children-St. Louis , St. Louis, Missouri
| | - Magali Cucchiarini
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University , Homburg/Saar, Germany
| |
Collapse
|
9
|
Negri T, Brich S, Bozzi F, Volpi CV, Gualeni AV, Stacchiotti S, De Cecco L, Canevari S, Gloghini A, Pilotti S. New transcriptional-based insights into the pathogenesis of desmoplastic small round cell tumors (DSRCTs). Oncotarget 2018; 8:32492-32504. [PMID: 28415643 PMCID: PMC5464804 DOI: 10.18632/oncotarget.16477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022] Open
Abstract
To gain new insights into desmoplastic small round cell tumors (DSRCTs) by means of gene expression profiling (GEP). Formalin-fixed, paraffin-embedded surgical specimens obtained from seven pretreated DSRCT patients were interrogated using GEP complemented by immunohistochemistry, a cancer stem cell array, and miRNA in situ hybridisation, including the combined chimera modules miRNA-200/ZEB1 and miRNA-34/SLUG. The chimera modules divided the cases into three classes that respectively recapitulated the traits of mesenchymal epithelial reverse transition (MErT), epithelial mesenchymal transition (EMT), and hybrid/partial EMT. This indicates a close correlation between the reprogramming governed by EMT regulators and DSRCT biology, which was further confirmed by miRNA-21 and is consistent with the broad morphological spectrum of DSRCTs. Starting from the miRNA-200/ZEB1 axis, we also found that DSRCTs carry a signature of immunological ignorance that is not responsive to PD-L1 blockade. Evidence that the up-regulation of miRNA-200 and E-cadherin, and quite a high level of miRNA-21 expression segregate with the MErT supports the idea that, in addition to the hybrid/partial state, MErT is also enriched in stemness: the androgen-positive cases, whose stemness traits were confirmed by stem cell arrays, all fell into these two classes. Our findings also confirmed that tumoral cell PDGFRA expression correlates with desmoplasia, and demonstrated the co-expression of PDGFRA and ISLR/Meflin, another marker of pluripotency. Despite the limited number of cases, these findings provide unexpectedly relevant information concerning the pathogenesis of DSRCTs, and prove the validity of miRNA-based chimera circuit modelling in the clinico-pathological setting.
Collapse
Affiliation(s)
- Tiziana Negri
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Brich
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,MOSE-DEA, University of Trieste, Trieste, Italy
| | - Fabio Bozzi
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara V Volpi
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ambra V Gualeni
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor and Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Canevari
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
10
|
Cho HJ, Lee JW, Cho HJ, Lee CS, Kim HS. Identification of Adult Mesodermal Progenitor Cells and Hierarchy in Atherosclerotic Vascular Calcification. Stem Cells 2018; 36:1075-1096. [PMID: 29484798 DOI: 10.1002/stem.2814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/26/2018] [Accepted: 02/05/2018] [Indexed: 01/01/2023]
Abstract
The nature of calcifying progenitor cells remains elusive. In this study, we investigated the developmental hierarchy and dynamics of progenitor cells. In vitro and in vivo reconstitution assays demonstrated that Sca-1+/PDGFRα- cells in the bone marrow (BM) are the ancestors of Sca-1+/PDGFRα+ cells. Cells of CD29 + Sca-1+/PDGFRα- lineage in the BM showed both hematopoietic potential with osteoclastic differentiation ability as well as mesenchymal stem cell-like properties with osteoblastic differentiation potential. Clonally-isolated BM-derived artery-infiltrated Sca-1+/PDGFRα- cells maintained osteoblastic/osteoclastic bipotency but lost hematopoietic activity. In hypercholesterolemic apolipoprotein-E-deficient (Apoe-/-) mice, the mobilization from BM to peripheral circulation, followed by migration into atherosclerotic plaques of Sca-1+/PDGFRα- cells, but not Sca-1+/PDGFRα+ cells, were significantly decreased, and Interleukin-1β (IL-1β) and Interleukin-5 (IL-5) mediated this response. Here, we demonstrated that Sca-1+/PDGFRα- cells are mesodermal progenitor cells in adults, and the dynamics of progenitor cells were regulated by atherosclerosis-related humoral factors. These results may contribute to better understanding of vascular homeostasis and assist in the development of novel therapies for atherosclerosis. Stem Cells 2018;36:1075-1096.
Collapse
Affiliation(s)
- Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin-Woo Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Ju Cho
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
| | - Choon-Soo Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea.,World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea.,World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
11
|
Bozzi F, Brich S, Dagrada GP, Negri T, Conca E, Cortelazzi B, Belfiore A, Perrone F, Gualeni AV, Gloghini A, Cabras A, Brenca M, Maestro R, Zaffaroni N, Casali P, Bertulli R, Deraco M, Pilotti S. Epithelioid peritoneal mesothelioma: a hybrid phenotype within a mesenchymal-epithelial/epithelial-mesenchymal transition framework. Oncotarget 2018; 7:75503-75517. [PMID: 27705913 PMCID: PMC5342756 DOI: 10.18632/oncotarget.12262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/13/2016] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to reconsider the biological characteristics of epithelioid malignant peritoneal mesothelioma (E-MpM) in the light of new concepts about epithelial mesenchymal transition and mesenchymal epithelial reverse transition (EMT/MErT) and the role of epigenetic reprogramming in this context. To this end we profiled surgical specimens and derived cells cultures by a number of complementary approaches i.e. immunohistochemistry, immunofluorescence, in situ hybridization, biochemistry, pluripotent stem cell arrays, treatments with cytokines, growth factors and specific inhibitors.The analyses of the surgical specimens showed that i) EZH2 is expressed throughout the spectrum of MpM, ii) that E-MpM (including the high-grade undifferentiated form) are characterised by c-MYC and miRNA 17-5p expression, and iii) that progression to sarcomatoid MpM is dictated by EMT regulators. They also showed that E-MpM expressed c-MET and are enriched in E- and P-cadherins- and VEGFR2-expressing CSCs, thus strongly supporting a role for MErT reprogramming in endowing E-MpM tumour cells with stemness and plasticity, and hence with a drug resistant phenotype. The cell culture-based experiments confirmed the stemness traits and plasticity of E-MpM, and support the view that EZH2 is a druggable target in this tumor.
Collapse
Affiliation(s)
- Fabio Bozzi
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Brich
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,MOSE-DEA University of Trieste, Trieste, Italy
| | - Gian Paolo Dagrada
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tiziana Negri
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Conca
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Cortelazzi
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonino Belfiore
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ambra Vittoria Gualeni
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonello Cabras
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Brenca
- Experimental Oncology 1, Centro di Riferimento Oncologico, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Roberta Maestro
- Experimental Oncology 1, Centro di Riferimento Oncologico, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Casali
- Adult Mesenchymal Tumor Medical Oncology Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rossella Bertulli
- Adult Mesenchymal Tumor Medical Oncology Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcello Deraco
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
12
|
Gonzalez ME, Martin EE, Anwar T, Arellano-Garcia C, Medhora N, Lama A, Chen YC, Tanager KS, Yoon E, Kidwell KM, Ge C, Franceschi RT, Kleer CG. Mesenchymal Stem Cell-Induced DDR2 Mediates Stromal-Breast Cancer Interactions and Metastasis Growth. Cell Rep 2017; 18:1215-1228. [PMID: 28147276 DOI: 10.1016/j.celrep.2016.12.079] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 11/28/2016] [Accepted: 12/22/2016] [Indexed: 12/18/2022] Open
Abstract
Increased collagen deposition by breast cancer (BC)-associated mesenchymal stem/multipotent stromal cells (MSC) promotes metastasis, but the mechanisms are unknown. Here, we report that the collagen receptor discoidin domain receptor 2 (DDR2) is essential for stromal-BC communication. In human BC metastasis, DDR2 is concordantly upregulated in metastatic cancer and multipotent mesenchymal stromal cells. In MSCs isolated from human BC metastasis, DDR2 maintains a fibroblastic phenotype with collagen deposition and induces pathological activation of DDR2 signaling in BC cells. Loss of DDR2 in MSCs impairs their ability to promote DDR2 phosphorylation in BC cells, as well as BC cell alignment, migration, and metastasis. Female ddr2-deficient mice homozygous for the slie mutation show inefficient spontaneous BC metastasis. These results point to a role for mesenchymal stem cell DDR2 in metastasis and suggest a therapeutic approach for metastatic BC.
Collapse
Affiliation(s)
- Maria E Gonzalez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily E Martin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Talha Anwar
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Caroline Arellano-Garcia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Natasha Medhora
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arjun Lama
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yu-Chih Chen
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin S Tanager
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kelley M Kidwell
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunxi Ge
- School of Dentistry, Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Renny T Franceschi
- School of Dentistry, Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Majidinia M, Sadeghpour A, Yousefi B. The roles of signaling pathways in bone repair and regeneration. J Cell Physiol 2017; 233:2937-2948. [DOI: 10.1002/jcp.26042] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Maryam Majidinia
- Solid Tumor Research Center; Urmia University of Medical Sciences; Urmia Iran
| | - Alireza Sadeghpour
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital; Tabriz University of Medical Sciences; Tabriz Iran
- Drug Applied Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Bahman Yousefi
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Molecular Targeting Therapy Research Group; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
- Stem cell and Regenerative Medicine Institute; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
14
|
Frisch J, Orth P, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Kohn D, Cucchiarini M. Peripheral blood aspirates overexpressing IGF-I via rAAV gene transfer undergo enhanced chondrogenic differentiation processes. J Cell Mol Med 2017; 21:2748-2758. [PMID: 28467017 PMCID: PMC5661259 DOI: 10.1111/jcmm.13190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/09/2017] [Indexed: 01/24/2023] Open
Abstract
Implantation of peripheral blood aspirates induced towards chondrogenic differentiation upon genetic modification in sites of articular cartilage injury may represent a powerful strategy to enhance cartilage repair. Such a single‐step approach may be less invasive than procedures based on the use of isolated or concentrated MSCs, simplifying translational protocols in patients. In this study, we provide evidence showing the feasibility of overexpressing the mitogenic and pro‐anabolic insulin‐like growth factor I (IGF‐I) in human peripheral blood aspirates via rAAV‐mediated gene transfer, leading to enhanced proliferative and chondrogenic differentiation (proteoglycans, type‐II collagen, SOX9) activities in the samples relative to control (reporter rAAV‐lacZ) treatment over extended periods of time (at least 21 days, the longest time‐point evaluated). Interestingly, IGF‐I gene transfer also triggered hypertrophic, osteo‐ and adipogenic differentiation processes in the aspirates, suggesting that careful regulation of IGF‐I expression may be necessary to contain these events in vivo. Still, the current results demonstrate the potential of targeting human peripheral blood aspirates via therapeutic rAAV transduction as a novel, convenient tool to treat articular cartilage injuries.
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Patrick Orth
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
15
|
Targeting the TAM Receptors in Leukemia. Cancers (Basel) 2016; 8:cancers8110101. [PMID: 27834816 PMCID: PMC5126761 DOI: 10.3390/cancers8110101] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/21/2016] [Accepted: 11/01/2016] [Indexed: 12/20/2022] Open
Abstract
Targeted inhibition of members of the TAM (TYRO-3, AXL, MERTK) family of receptor tyrosine kinases has recently been investigated as a novel strategy for treatment of hematologic malignancies. The physiologic functions of the TAM receptors in innate immune control, natural killer (NK) cell differentiation, efferocytosis, clearance of apoptotic debris, and hemostasis have previously been described and more recent data implicate TAM kinases as important regulators of erythropoiesis and megakaryopoiesis. The TAM receptors are aberrantly or ectopically expressed in many hematologic malignancies including acute myeloid leukemia, B- and T-cell acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma. TAM receptors contribute to leukemic phenotypes through activation of pro-survival signaling pathways and interplay with other oncogenic proteins such as FLT3, LYN, and FGFR3. The TAM receptors also contribute to resistance to both cytotoxic chemotherapeutics and targeted agents, making them attractive therapeutic targets. A number of translational strategies for TAM inhibition are in development, including small molecule inhibitors, ligand traps, and monoclonal antibodies. Emerging areas of research include modulation of TAM receptors to enhance anti-tumor immunity, potential roles for TYRO-3 in leukemogenesis, and the function of the bone marrow microenvironment in mediating resistance to TAM inhibition.
Collapse
|
16
|
Manning LB, Li Y, Chickmagalur NS, Li X, Xu L. Discoidin Domain Receptor 2 as a Potential Therapeutic Target for Development of Disease-Modifying Osteoarthritis Drugs. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3000-3010. [PMID: 27640147 DOI: 10.1016/j.ajpath.2016.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/26/2016] [Accepted: 06/30/2016] [Indexed: 02/05/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis disorders, but the identification of therapeutic targets to effectively prevent OA has been increasingly difficult. The goal of this investigation is to provide experimental evidence that discoidin domain receptor 2 (DDR2) may be an ideal target for the development of disease-modifying OA drugs. Ddr2 was conditionally deleted from articular cartilage of adult mouse knee joints. Aggrecan-CreERT2;floxed Ddr2 mice, which were generated by crossing Aggrecan-CreERT2 mice with floxed Ddr2 mice, then received tamoxifen injections at the age of 8 weeks. The mice were then subjected to destabilization of the medial meniscus (DMM) surgery. At 8 and 16 weeks after DMM, mice were euthanized for the collection of knee joints. In a separate experiment, Aggrecan-CreERT2;floxed Ddr2 mice were subjected to DMM at the age of 10 weeks. The mice then received tamoxifen injections at 8 weeks after DMM. The mice were euthanized for the collection of knee joints at 16 weeks after DMM. The progressive process of articular cartilage degeneration was significantly delayed in the knee joints of Ddr2-deficient mice in comparison to their control littermates. Articular cartilage damage in the knee joints of the mice was associated with increased expression profiles of both Ddr2 and matrix metalloproteinase 13. These findings suggest that DDR2 may be an ideal target for the development of disease-modifying OA drugs.
Collapse
Affiliation(s)
- Lauren B Manning
- Department of Prosthodontics, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Yefu Li
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Nithya S Chickmagalur
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Xiaolong Li
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Ferluga S, Tomé CML, Herpai DM, D'Agostino R, Debinski W. Simultaneous targeting of Eph receptors in glioblastoma. Oncotarget 2016; 7:59860-59876. [PMID: 27494882 PMCID: PMC5312354 DOI: 10.18632/oncotarget.10978] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/16/2016] [Indexed: 12/20/2022] Open
Abstract
Eph tyrosine kinase receptors are frequently overexpressed and functional in many cancers, and they are attractive candidates for targeted therapy. Here, we analyzed the expression of Eph receptor A3, one of the most up-regulated factors in glioblastoma cells cultured under tumorsphere-forming conditions, together with EphA2 and EphB2 receptors. EphA3 was overexpressed in up to 60% of glioblastoma tumors tested, but not in normal brain. EphA3 was localized in scattered areas of the tumor, the invasive ring, and niches near tumor vessels. EphA3 co-localized with macrophage/leukocyte markers, suggesting EphA3 expression on tumor-infiltrating cells of bone marrow origin. We took advantage of the fact that ephrinA5 (eA5) is a ligand that binds EphA3, EphA2 and EphB2 receptors, and used it to construct a novel targeted anti-glioblastoma cytotoxin. The eA5-based cytotoxin potently and specifically killed glioblastoma cells with an IC50 of at least 10-11 M. This and similar cytotoxins will simultaneously target different compartments of glioblastoma tumors while mitigating tumor heterogeneity.
Collapse
Affiliation(s)
- Sara Ferluga
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Carla Maria Lema Tomé
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Denise Mazess Herpai
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Section on Biostatistics, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | - Waldemar Debinski
- Department of Cancer Biology, Radiation Oncology and Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
18
|
Frisch J, Orth P, Venkatesan JK, Rey‐Rico A, Schmitt G, Kohn D, Madry H, Cucchiarini M. Genetic Modification of Human Peripheral Blood Aspirates Using Recombinant Adeno-Associated Viral Vectors for Articular Cartilage Repair with a Focus on Chondrogenic Transforming Growth Factor-β Gene Delivery. Stem Cells Transl Med 2016; 6:249-260. [PMID: 28170175 PMCID: PMC5442727 DOI: 10.5966/sctm.2016-0149] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/28/2016] [Indexed: 01/13/2023] Open
Abstract
Transplantation of genetically modified peripheral blood aspirates that carry chondrogenically competent progenitor cells may offer new, convenient tools to treat articular cartilage lesions compared with the more complex and invasive application of bone marrow concentrates or of bone marrow‐derived mesenchymal stem cells. Here, we show that recombinant adeno‐associated viral (rAAV) vectors are powerful gene vehicles capable of successfully targeting primary human peripheral blood aspirates in a stable and safe manner, allowing for an efficient and long‐term transgene expression in such samples (up to 63 days with use of a lacZ reporter gene and for at least 21 days with application of the pleiotropic, chondrogenic factor transforming growth factor‐β [TGF‐β]). rAAV‐mediated overexpression of TGF‐β enhanced both the proliferative and metabolic properties of the peripheral blood aspirates, also increasing the chondrogenic differentiation processes in these samples. Hypertrophy and osteogenic differentiation events were also activated by production of TGF‐β via rAAV, suggesting that translation of the current approach in vivo will probably require close regulation of expression of this candidate gene. However, these results support the concept of directly modifying peripheral blood as a novel approach to conveniently treat articular cartilage lesions in patients. Stem Cells Translational Medicine2017;6:249–260
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Patrick Orth
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Ana Rey‐Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
19
|
Osteogenic signaling on silk-based matrices. Biomaterials 2016; 97:133-53. [DOI: 10.1016/j.biomaterials.2016.04.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/25/2016] [Accepted: 04/20/2016] [Indexed: 12/11/2022]
|
20
|
Frisch J, Venkatesan JK, Rey-Rico A, Zawada AM, Schmitt G, Madry H, Cucchiarini M. Effects of rAAV-mediated FGF-2 gene transfer and overexpression upon the chondrogenic differentiation processes in human bone marrow aspirates. J Exp Orthop 2016; 3:16. [PMID: 27473203 PMCID: PMC4967065 DOI: 10.1186/s40634-016-0052-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/22/2016] [Indexed: 12/31/2022] Open
Abstract
Background Application of genetically modified bone marrow concentrates in articular cartilage lesions is a promising approach to enhance cartilage repair by stimulating the chondrogenic differentiation processes in sites of injury. Method In the present study, we examined the potential benefits of transferring the proliferative and pro-chondrogenic basic fibroblast growth factor (FGF-2) to human bone marrow aspirates in vitro using the clinically adapted recombinant adeno-associated virus (rAAV) vectors to monitor the biological and chondrogenic responses over time to the treatment compared with control (lacZ) gene application. Results Effective, significant FGF-2 gene transfer and expression via rAAV was established in the aspirates relative to the lacZ condition (from ~ 97 to 36 pg rhFGF-2/mg total proteins over an extended period of 21 days). Administration of the candidate FGF-2 vector led to prolonged increases in cell proliferation, matrix synthesis, and chondrogenesis but also to hypertrophic and terminal differentiation in the aspirates. Conclusion The present evaluation shows the advantages of rAAV-mediated FGF-2 gene transfer to conveniently modify bone marrow concentrates as a future approach to directly treat articular cartilage lesions, provided that expression of the growth factor is tightly regulated to prevent premature hypertrophy in vivo.
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Adam M Zawada
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany.
| |
Collapse
|
21
|
Lombardi R, Chen SN, Ruggiero A, Gurha P, Czernuszewicz GZ, Willerson JT, Marian AJ. Cardiac Fibro-Adipocyte Progenitors Express Desmosome Proteins and Preferentially Differentiate to Adipocytes Upon Deletion of the Desmoplakin Gene. Circ Res 2016; 119:41-54. [PMID: 27121621 DOI: 10.1161/circresaha.115.308136] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/27/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Mutations in desmosome proteins cause arrhythmogenic cardiomyopathy (AC), a disease characterized by excess myocardial fibroadipocytes. Cellular origin(s) of fibroadipocytes in AC is unknown. OBJECTIVE To identify the cellular origin of adipocytes in AC. METHODS AND RESULTS Human and mouse cardiac cells were depleted from myocytes and flow sorted to isolate cells expressing platelet-derived growth factor receptor-α and exclude those expressing other lineage and fibroblast markers (CD32, CD11B, CD45, Lys76, Ly(-6c) and Ly(6c), thymocyte differentiation antigen 1, and discoidin domain receptor 2). The PDGFRA(pos):Lin(neg):THY1(neg):DDR2(neg) cells were bipotential as the majority expressed collagen 1 α-1, a fibroblast marker, and a subset CCAAT/enhancer-binding protein α, a major adipogenic transcription factor, and therefore, they were referred to as fibroadipocyte progenitors (FAPs). FAPs expressed desmosome proteins, including desmoplakin, predominantly in the adipogenic but not fibrogenic subsets. Conditional heterozygous deletion of Dsp in mice using Pdgfra-Cre deleter led to increased fibroadipogenesis in the heart and mild cardiac dysfunction. Genetic fate mapping tagged 41.4±4.1% of the cardiac adipocytes in the Pdgfra-Cre:Eyfp:Dsp(W/F) mice, indicating an origin from FAPs. FAPs isolated from the Pdgfra-Cre:Eyfp:Dsp(W/F) mouse hearts showed enhanced differentiation to adipocytes. Mechanistically, deletion of Dsp was associated with suppressed canonical Wnt signaling and enhanced adipogenesis. In contrast, activation of the canonical Wnt signaling rescued adipogenesis in a dose-dependent manner. CONCLUSIONS A subset of cardiac FAPs, identified by the PDGFRA(pos):Lin(neg):THY1(neg):DDR2(neg) signature, expresses desmosome proteins and differentiates to adipocytes in AC through a Wnt-dependent mechanism. The findings expand the cellular spectrum of AC, commonly recognized as a disease of cardiac myocytes, to include nonmyocyte cells in the heart.
Collapse
Affiliation(s)
- Raffaella Lombardi
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.).
| | - Suet Nee Chen
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.)
| | - Alessandra Ruggiero
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.)
| | - Priyatansh Gurha
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.)
| | - Grazyna Z Czernuszewicz
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.)
| | - James T Willerson
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.)
| | - Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Science Center at Houston (R.L., S.N.C., A.R., P.G., G.Z.C., A.J.M.); and Department of Medicine, Texas Heart Institute, Houston (J.T.W.).
| |
Collapse
|
22
|
Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. TGF-β gene transfer and overexpression via rAAV vectors stimulates chondrogenic events in human bone marrow aspirates. J Cell Mol Med 2016; 20:430-40. [PMID: 26808466 PMCID: PMC4759465 DOI: 10.1111/jcmm.12774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
Genetic modification of marrow concentrates may provide convenient approaches to enhance the chondrogenic differentiation processes and improve the repair capacities in sites of cartilage defects following administration in the lesions. Here, we provided clinically adapted recombinant adeno‐associated virus (rAAV) vectors to human bone marrow aspirates to promote the expression of the potent transforming growth factor beta (TGF‐β) as a means to regulate the biological and chondrogenic activities in the samples in vitro. Successful TGF‐β gene transfer and expression viarAAV was reached relative to control (lacZ) treatment (from 511.1 to 16.1 pg rhTGF‐β/mg total proteins after 21 days), allowing to durably enhance the levels of cell proliferation, matrix synthesis, and chondrogenic differentiation. Strikingly, in the conditions applied here, application of the candidate TGF‐β vector was also capable of reducing the hypertrophic and osteogenic differentiation processes in the aspirates, showing the potential benefits of using this particular vector to directly modify marrow concentrates to generate single‐step, effective approaches that aim at improving articular cartilage repair in vivo.
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
23
|
Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. Chondrogenic Differentiation Processes in Human Bone Marrow Aspirates upon rAAV-Mediated Gene Transfer and Overexpression of the Insulin-Like Growth Factor I. Tissue Eng Part A 2015; 21:2460-71. [PMID: 26123891 DOI: 10.1089/ten.tea.2014.0679] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Direct therapeutic gene transfer in marrow concentrates is an attractive strategy to conveniently enhance the chondrogenic differentiation processes as a means to improve the healing response of damaged articular cartilage upon reimplantation in sites of injury. In the present study, we evaluated the ability of the clinically adapted recombinant adeno-associated virus (rAAV) vectors to mediate overexpression of the insulin-like growth factor I (IGF-I) in human bone marrow aspirates that may modulate the proliferative, anabolic activities, and chondrogenic differentiation potential in such samples in vitro. The results demonstrate that successful, significant rAAV-mediated IGF-I gene transfer and expression were achieved in transduced aspirates (up to 105.9±35.1 pg rhIGF-I/mg total proteins) over time (21 days) at very high levels (∼80% of cells expressing the candidate IGF-I transgene), leading to increased levels of proliferation, matrix synthesis, and chondrogenic differentiation over time compared with the control (lacZ) condition. Treatment with the candidate IGF-I vector also stimulated the hypertrophic and osteogenic differentiation processes in the aspirates, suggesting that the regulation of IGF-I expression through rAAV will be a prerequisite for future translation of the approach in vivo. However, these findings show the possible benefits of this vector class to directly modify marrow concentrates as a convenient tool for strategies that aim at improving the repair of articular cartilage lesions.
Collapse
Affiliation(s)
- Janina Frisch
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg/Saar, Germany
| | - Ana Rey-Rico
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg/Saar, Germany
| | | | - Gertrud Schmitt
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg/Saar, Germany
| | - Henning Madry
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg/Saar, Germany .,2 Department of Orthopedic Surgery, Saarland University Medical Center , Homburg/Saar, Germany
| | - Magali Cucchiarini
- 1 Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg/Saar, Germany
| |
Collapse
|
24
|
Abstract
The control of cellular growth and proliferation is key to the maintenance of homeostasis. Survival, proliferation, and arrest are regulated, in part, by Growth Arrest Specific 6 (Gas6) through binding to members of the TAM receptor tyrosine kinase family. Activation of the TAM receptors leads to downstream signaling through common kinases, but the exact mechanism within each cellular context varies and remains to be completely elucidated. Deregulation of the TAM family, due to its central role in mediating cellular proliferation, has been implicated in multiple diseases. Axl was cloned as the first TAM receptor in a search for genes involved in the progression of chronic to acute-phase leukemia, and has since been established as playing a critical role in the progression of cancer. The oncogenic nature of Axl is demonstrated through its activation of signaling pathways involved in proliferation, migration, inhibition of apoptosis, and therapeutic resistance. Despite its recent discovery, significant progress has been made in the development of effective clinical therapeutics targeting Axl. In order to accurately define the role of Axl in normal and diseased processes, it must be analyzed in a cell type-specific context.
Collapse
|
25
|
Hayrapetyan A, Jansen JA, van den Beucken JJJP. Signaling pathways involved in osteogenesis and their application for bone regenerative medicine. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:75-87. [PMID: 25015093 DOI: 10.1089/ten.teb.2014.0119] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bone regeneration is a well organized but complex physiological process, in which different cell types and their activated signaling pathways are involved. In bone regeneration and remodeling processes, mesenchymal stem cells (MSCs) have a crucial role, and their differentiation during these processes is regulated by specific signaling molecules (growth factors/cytokines and hormones) and their activated intracellular networks. Especially the utilization of the molecular machinery seems crucial to consider prior to developing bone implants, bone-substitute materials, and cell-based constructs for bone regeneration. The aim of this review is to provide an overview of the signaling mechanisms involved in bone regeneration and remodeling and the osteogenic potential of MSCs to become a key cellular resource for such regeneration and remodeling processes. Additionally, an overview of possibilities to beneficially exploit cell signaling processes to optimize bone regeneration is provided.
Collapse
|