1
|
Cai L, Kar P, Liu Y, Chu X, Sharma A, Lee TJ, Arbab A, Raju RP. Plasma Extracellular Vesicle-Derived miR-296-5p is a Maturation-Dependent Rejuvenation Factor that Downregulates Inflammation and Improves Survival after Sepsis. J Extracell Vesicles 2025; 14:e70065. [PMID: 40285735 PMCID: PMC12032680 DOI: 10.1002/jev2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
There is a progressive decline in physiological function with age, and aging is associated with increased susceptibility to injury and infection. However, several reports have indicated that the agility of youth is characterized by transferable rejuvenating molecular factors, as was observed previously in heterochronic parabiosis experiments. These experiments demonstrated a rejuvenating effect of young blood in old animals. There have been several efforts to characterize these youthful or maturation-associated factors in the young blood. In this report, we demonstrate the resilience of young mice, at or before puberty, to polymicrobial sepsis and show an age-dependent effect of small extracellular vesicles (EVs) from plasma on the outcome following sepsis. The EVs from the young mice were cytoprotective, anti-inflammatory, and reduced cellular senescence markers. MicroRNA sequencing of the EVs showed an age-associated signature and identified miR-296-5p and miR-541-5p to progressively reduce their levels in the blood plasma with increasing age. We further show that the levels of these miRNAs decline with age in multiple organs. The miRNAs miR-296-5p and miR-541-5p showed a reparatory effect in an in vitro wound healing model and the miR-296-5p, when given intraperitoneally, reduced mortality in the mouse model of sepsis. In summary, our studies demonstrate that EVs from very young mice have a reparative effect on sepsis, and the reparative factors are likely maturation-dependent. Our observation that miR-296-5p and miR-541-5p are plasma EV constituents that significantly reduce with age and can reduce inflammation suggests a therapeutic potential for these miRNAs in inflammation and age-associated diseases.
Collapse
Affiliation(s)
- Lun Cai
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Parmita Kar
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Yutao Liu
- Department of Cell Biology and AnatomyMedical College of GeorgiaAugustaGeorgiaUSA
| | - Xiaogang Chu
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Ashok Sharma
- Center for Biotechnology and Genomic MedicineMedical College of GeorgiaAugustaGeorgiaUSA
| | - Tae Jin Lee
- Center for Biotechnology and Genomic MedicineMedical College of GeorgiaAugustaGeorgiaUSA
| | - Ali Arbab
- Georgia Cancer CenterMedical College of GeorgiaAugustaGeorgiaUSA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
- Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| |
Collapse
|
2
|
Kaviyaprabha R, Miji TV, Suseela R, Muthusami S, Thangaleela S, Almoallim HS, Sivakumar P, Bharathi M. Screening miRNAs to Hinder the Tumorigenesis of Renal Clear Cell Carcinoma Associated with KDR Expression. Curr Cancer Drug Targets 2025; 25:183-203. [PMID: 39289946 DOI: 10.2174/0115680096321287240826065718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION This study delved to understand the role of Kinase Insert Domain Receptor (KDR) and its associated miRNAs in renal cell carcinoma through an extensive computational analysis. The potential of our findings to guide future research in this area is significant. METHODS Our methods, which included the use of UALCAN and GEPIA2 databases, as well as miRDB, MirDIP, miRNet v2.0, miRTargetLink, MiEAA v2.1, TarBase v8.0, INTERNET, and miRTarBase, were instrumental in identifying the regulation of miRNA associated with KDR expression. The predicted miRNA was validated with the TCGA-KIRC patients' samples by implementing CancerMIRNome. The TargetScanHuman v8.0 was implemented to identify the associations between human miRNAs and KDR. A Patch Dock server analyzed the interactions between hsa-miR-200c-3p and KDR. RESULTS The KDR expression rate was investigated in the Kidney Renal Cell Carcinoma (KIRC) samples, and adjacent normal tissues revealed that the expression rate was significantly higher than the normal samples, which was evident from the strong statistical significance (P = 1.63e-12). Likely, the KDR expression rate was estimated as high at tumor grade 1 and gradually decreased till the metastasis grade, reducing the survival rate of the KIRC patients. To identify these signals early, we predicted a miRNA that could alter the expression of KDR. Furthermore, we uncovered the potential associations between miR-200c-3p expressions by regulating KDR towards the progression of KIRC. DISCUSSION Upon examining the outcome, it became evident that miR-200c-3p was significantly downregulated in KIRC compared to the normal samples. Moreover, the negative correlation was obtained for hsa-miR-200c-3p (R = - 0.276) along with the KDR expression describing that the increased rate of hsamiR- 200c-3p might reduce the KDR expression rate, which may suppress the KIRC initiation or progression. CONCLUSION The in-silico analysis indicated that the significant increase in KDR expression during the initiation of KIRC could serve as an early diagnostic marker. Moreover, KDR could be utilized to identify advancements in KIRC stages. Additionally, hsa-miR-200c-3p was identified as a potential regulator capable of downregulating and upregulating KDR expression among the 24 miRNAs screened. This finding holds promise for future research endeavors. Concurrent administration of the FDA-approved 5- fluorouracil with KIRC drugs, such as sorafenib, zidovudine, and everolimus, may have the potential to enhance the therapeutic efficacy in downregulating hsa-miR-200c-3p. However, further in vitro studies are imperative to validate these findings and gain a comprehensive understanding of the intricate regulatory interplay involving hsa-miR-200c-3p, KDR, 5-fluorouracil, and other FDA-approved drugs for the treatment of KIRC. This will facilitate the identification of KIRC stage progression and its underlying preventative mechanisms.
Collapse
Affiliation(s)
- Rangaraj Kaviyaprabha
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Thandaserry Vasudevan Miji
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Rangaraj Suseela
- Centre for Cancer Research, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Sridhar Muthusami
- Centre for Cancer Research, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Subramanian Thangaleela
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602 105, Tamil Nadu, India
| | - Hesham S Almoallim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, King Saud University, PO Box-60169, Riyadh -11545, Saudi Arabia
| | - Priyadarshini Sivakumar
- Department of Microbiology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| |
Collapse
|
3
|
Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci 2024; 25:2356. [PMID: 38397032 PMCID: PMC10889096 DOI: 10.3390/ijms25042356] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Adipose tissue (AT) is a large and important energy storage organ as well as an endocrine organ with a critical role in many processes. Additionally, AT is an enormous and easily accessible source of multipotent cell types used in our day for all types of tissue regeneration. The ability of adipose-derived stem cells (ADSCs) to differentiate into other types of cells, such as endothelial cells (ECs), vascular smooth muscle cells, or cardiomyocytes, is used in tissue engineering in order to promote/stimulate the process of angiogenesis. Being a key for future successful clinical applications, functional vascular networks in engineered tissue are targeted by numerous in vivo and ex vivo studies. The article reviews the angiogenic potential of ADSCs and explores their capacity in the field of tissue engineering (TE).
Collapse
Affiliation(s)
- Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Toronto, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada
| |
Collapse
|
4
|
Tossetta G, Piani F, Borghi C, Marzioni D. Role of CD93 in Health and Disease. Cells 2023; 12:1778. [PMID: 37443812 PMCID: PMC10340406 DOI: 10.3390/cells12131778] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
CD93 (also known as complement protein 1 q subcomponent receptor C1qR1 or C1qRp), is a transmembrane glycoprotein encoded by a gene located on 20p11.21 and composed of 652 amino acids. CD93 can be present in two forms: soluble (sCD93) and membrane-bound (CD93). CD93 is mainly expressed on endothelial cells, where it plays a key role in promoting angiogenesis both in physiology and disease, such as age-related macular degeneration and tumor angiogenesis. In fact, CD93 is highly expressed in tumor-associated vessels and its presence correlates with a poor prognosis, poor immunotherapy response, immune cell infiltration and high tumor, node and metastasis (TNM) stage in many cancer types. CD93 is also expressed in hematopoietic stem cells, cytotrophoblast cells, platelets and many immune cells, i.e., monocytes, neutrophils, B cells and natural killer (NK) cells. Accordingly, CD93 is involved in modulating important inflammatory-associated diseases including systemic sclerosis and neuroinflammation. Finally, CD93 plays a role in cardiovascular disease development and progression. In this article, we reviewed the current literature regarding the role of CD93 in modulating angiogenesis, inflammation and tumor growth in order to understand where this glycoprotein could be a potential therapeutic target and could modify the outcome of the abovementioned pathologies.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Federica Piani
- Cardiovascular Medicine Unit, Heart, Chest and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (C.B.)
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Claudio Borghi
- Cardiovascular Medicine Unit, Heart, Chest and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (C.B.)
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| |
Collapse
|
5
|
Zhu D, Guo T, Xu J, Yuan D, Lin M, Yang M. Elevated Expression of miR-296 in Human Placentas and Serum Samples From Pregnancies With Preeclampsia. Br J Biomed Sci 2023; 80:11004. [PMID: 37113621 PMCID: PMC10126236 DOI: 10.3389/bjbs.2023.11004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/07/2023] [Indexed: 04/29/2023]
Abstract
Background: Preeclampsia (PE) is a hypertensive disorder of pregnancy characterized by widespread maternal endothelial dysfunction. Although clinical signs subside following delivery, long-term risks associated with PE include hypertension, stroke, and cardiovascular disease. MicroRNAs (miRNAs) are emerging as critical regulators of biological function, and while alterations to the miRNAs have been described in the context of pregnancy and PE, the postpartum implications of PE on miRNA expression are unknown. In the present study, we aimed to determine the clinical performance of miR-296 in PE. Methods: First, the clinical information and outcomes of all the participants were collected and analyzed. Afterward, the miR-296 expressions in the serum samples from healthy pregnant women and women with PE at different periods were detected using quantitative real-time polymerase chain reaction (qRT-PCR). Then, the receive operation characteristic (ROC) curve was used to determine the diagnostic value of miR-296 in PE. Finally, the at-term placentals were collected, the expressions of miR-296 in different groups were compared at first blood collection and at delivery. Results: In this study, we found that miR-296 expression was significantly increased in the placenta samples from PE patients compared with that in healthy controls both in early onset group (EOPE, p < 0.01) and late onset group (LOPE, p < 0.01). Furthermore, results of ROC analysis showed miR-296 might be a putative biomarker for early onset preeclampsia and late onset preeclampsia diagnosis with an area under the curve (AUC) of 0.84 (95% confidence interval 0.75-0.92) and 0.85 (95% confidence interval 0.77-0.93). Last but not the least, the expressions of miR-296 were significantly increased (p < 0.05) in serum samples of EOPE and LOPE patients (p < 0.001), and serum and placental levels of the miR-296 was positively correlated for EOPE (r = 0.5574, p < 0.001) and LOPE (r = 0.6613, p < 0.001) patients, respectively. Meanwhile, compared with those at first blood collection, the expression of miR-296 in EOPE (p = 0.05) and LOPE (p = 0.01) were significantly decreased at delivery. Conclusion: miR-296 may function as a putative diagnostic biomarker for PE and contribute to identifying at-risk mothers in pregnancy.
Collapse
Affiliation(s)
- Dandan Zhu
- The Department of Obstetrics and Gynecology, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
| | - Ting Guo
- Institute of Clinical Medicine, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
| | - Jie Xu
- Institute of Clinical Medicine, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
| | - Donglan Yuan
- The Department of Obstetrics and Gynecology, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
| | - Mei Lin
- Department of Clinical Laboratory, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
| | - Minyan Yang
- The Department of Obstetrics and Gynecology, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, China
- *Correspondence: Minyan Yang,
| |
Collapse
|
6
|
Magouliotis DE, Fergadi MP, Christodoulidis G, Svokos AA, Svokos KA, Bareka M, Athanasiou T. In-depth bioinformatic study of the cadherin 5 interactome in patients with thoracic aortic aneurysm unveils 8 novel biomarkers. Eur J Cardiothorac Surg 2021; 61:11-18. [PMID: 34293135 DOI: 10.1093/ejcts/ezab338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/25/2021] [Accepted: 06/12/2021] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVES Thoracic aortic aneurysm (TAA) is characterized by the dilation of the aorta and is associated with poor prognosis if not diagnosed and treated early. In this context, the identification of biomarkers regarding the TAA diagnosis, monitoring and prognosis is crucial. The purpose of the current study was to investigate the differential gene expression profile of the cadherin 5 (CDH5 or VE-Cadherin) gene network in patients with TAA, to propose novel biomarkers. METHODS In silico techniques were used to construct the interactome of the CDH5 network, identify the differentially expressed genes (DEGs) in TAA as compared to healthy controls, and uncover the related molecular functions and the regulating miRNAs. RESULTS Transcriptomic data of one microarray dataset were included, incorporating 43 TAA and 43 control samples. Eight DEGs were identified; 7 were up-regulated and 1 was down-regulated. A molecular signature of 8 genes (CDH5; Calcitonin Receptor-Like Receptor-CALCRL; Activin A Receptor-Like Type 1-ACVRL1, Tryptophanyl-TRNA Synthetase 1-WARS; Junction Plakoglobin-JUP, Protein Tyrosine Phosphatase Receptor Type J-PTPRJ, Purinergic Receptor P2X 4-P2RX4, Kinase Insert Domain Receptor-KDR) were identified as biomarkers associated with TAA. PTPRJ was associated with excellent discrimination and calibration in predicting TAA presentation. Positive correlations were reported regarding the expression of CDH5-CALCRL, CDH5-ACVRL1, CDH5-WARS and CDH5-PTPRJ. Finally, gene set enrichment analysis indicated the molecular functions and miRNA families (hsa-miR-296-5p, hsa-miR-6836-5p, hsa-miR-6132, hsa-miR-27a-5p and hsa-miR-6773-5p) relevant to the 8 biomarkers. CONCLUSIONS These outcomes propose an 8-gene molecular panel associated with TAA.
Collapse
Affiliation(s)
- Dimitrios E Magouliotis
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, UCL, London, UK
- Department of Cardiothoracic Surgery, University of Thessaly, Biopolis, Larissa, Greece
| | - Maria P Fergadi
- Department of Radiology, University of Thessaly, Biopolis, Larissa, Greece
| | | | - Alexis A Svokos
- Department of Obstetrics and Gynecology, Geisinger Medical Center, Danville, PA, USA
| | - Konstantina A Svokos
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Metaxia Bareka
- Department of Anesthesiology, University of Thessaly, Biopolis, Larissa, Greece
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| |
Collapse
|
7
|
Wang T, Guo Y, Liu S, Zhang C, Cui T, Ding K, Wang P, Wang X, Wang Z. KLF4, a Key Regulator of a Transitive Triplet, Acts on the TGF-β Signaling Pathway and Contributes to High-Altitude Adaptation of Tibetan Pigs. Front Genet 2021; 12:628192. [PMID: 33936161 PMCID: PMC8082500 DOI: 10.3389/fgene.2021.628192] [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: 11/11/2020] [Accepted: 03/10/2021] [Indexed: 11/13/2022] Open
Abstract
Tibetan pigs are native mammalian species on the Tibetan Plateau that have evolved distinct physiological traits that allow them to tolerate high-altitude hypoxic environments. However, the genetic mechanism underlying this adaptation remains elusive. Here, based on multitissue transcriptional data from high-altitude Tibetan pigs and low-altitude Rongchang pigs, we performed a weighted correlation network analysis (WGCNA) and identified key modules related to these tissues. Complex network analysis and bioinformatics analysis were integrated to identify key genes and three-node network motifs. We found that among the six tissues (muscle, liver, heart, spleen, kidneys, and lungs), lung tissue may be the key organs for Tibetan pigs to adapt to hypoxic environment. In the lung tissue of Tibetan pigs, we identified KLF4, BCL6B, EGR1, EPAS1, SMAD6, SMAD7, KDR, ATOH8, and CCN1 genes as potential regulators of hypoxia adaption. We found that KLF4 and EGR1 genes might simultaneously regulate the BCL6B gene, forming a KLF4-EGR1-BCL6B complex. This complex, dominated by KLF4, may enhance the hypoxia tolerance of Tibetan pigs by mediating the TGF-β signaling pathway. The complex may also affect the PI3K-Akt signaling pathway, which plays an important role in angiogenesis caused by hypoxia. Therefore, we postulate that the KLF4-EGR1-BCL6B complex may be beneficial for Tibetan pigs to survive better in the hypoxia environments. Although further molecular experiments and independent large-scale studies are needed to verify our findings, these findings may provide new details of the regulatory architecture of hypoxia-adaptive genes and are valuable for understanding the genetic mechanism of hypoxic adaptation in mammals.
Collapse
Affiliation(s)
- Tao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| | - Yuanyuan Guo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| | - Shengwei Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| | - Chaoxin Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| | - Tongyan Cui
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| | - Kun Ding
- College of Computer Science and Technology, Inner Mongolia Normal University, Hohhot, China
| | - Peng Wang
- HeiLongJiang Provincial Husbandry Department, Harbin, China
| | - Xibiao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhipeng Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Bioinformatics Center, Northeast Agricultural University, Harbin, China
| |
Collapse
|
8
|
Rahimian N, Razavi ZS, Aslanbeigi F, Mirkhabbaz AM, Piroozmand H, Shahrzad MK, Hamblin MR, Mirzaei H. Non-coding RNAs related to angiogenesis in gynecological cancer. Gynecol Oncol 2021; 161:896-912. [PMID: 33781555 DOI: 10.1016/j.ygyno.2021.03.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
Gynecological cancer affects the female reproductive system, including ovarian, uterine, endometrial, cervical, vulvar, and vaginal tumors. Non-coding RNAs (ncRNAs), and in particular microRNAs, function as regulatory molecules, which can control gene expression in a post-transcriptional manner. Normal physiological processes like cellular proliferation, differentiation, and apoptosis, and pathological processes such as oncogenesis and metastasis are regulated by microRNAs. Numerous reports have shown a direct role of microRNAs in the modulation of angiogenesis in gynecological cancer, via targeting pro-angiogenic factors and signaling pathways. Understanding the molecular mechanism involved in the regulation of angiogenesis by microRNAs may lead to new treatment options. Recently the regulatory role of some long non-coding RNAs in gynecological cancer has also been explored, but the information on this function is more limited. The aim of this article is to explore the pathways responsible for angiogenesis, and to what extent ncRNAs may be employed as biomarkers or therapeutic targets in gynecological cancer.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | | | | | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Karim Shahrzad
- Department of Internal Medicine and endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
9
|
Microvesicles from Human Immortalized Cell Lines of Endothelial Progenitor Cells and Mesenchymal Stem/Stromal Cells of Adipose Tissue Origin as Carriers of Bioactive Factors Facilitating Angiogenesis. Stem Cells Int 2020; 2020:1289380. [PMID: 32612661 PMCID: PMC7312709 DOI: 10.1155/2020/1289380] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/14/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022] Open
Abstract
Endothelial progenitor cells (EPCs) and mesenchymal stem/stromal cells (MSCs) are associated with maintaining tissue homeostasis and tissue repair. Both types of cells contribute to tissue regeneration through the secretion of trophic factors (alone or in the form of microvesicles). This study investigated the isolation and biological properties of microvesicles (MVs) derived from human immortalized MSC line HATMSC1 of adipose tissue origin and EPC line. The human immortalized cell line derived from the adipose tissue of a patient with venous stasis was established in our laboratory using the hTERT and pSV402 plasmids. The human EPC line originating from cord blood (HEPC-CB.1) was established in our previous studies. Microvesicles were isolated through a sequence of centrifugations. Analysis of the protein content of both populations of microvesicles, using the Membrane-Based Antibody Array and Milliplex ELISA showed that isolated microvesicles transported growth factors and pro- and antiangiogenic factors. Analysis of the miRNA content of isolated microvesicles revealed the presence of proangiogenic miRNA (miR-126, miR-296, miR-378, and miR-210) and low expression of antiangiogenic miRNA (miR-221, miR-222, and miR-92a) using real-time RT-PCR with the TaqMan technique. The isolated microvesicles were assessed for their effect on the proliferation and proangiogenic properties of cells involved in tissue repair. It was shown that both HEPC-CB.1- and HATMSC1-derived microvesicles increased the proliferation of human endothelial cells of dermal origin and that this effect was dose-dependent. In contrast, microvesicles had a limited impact on the proliferation of fibroblasts and keratinocytes. Both types of microvesicles improved the proangiogenic properties of human dermal endothelial cells, and this effect was also dose-dependent, as shown in the Matrigel assay. These results confirm the hypothesis that microvesicles of HEPC-CB.1 and HATMSC1 origin carry proteins and miRNAs that support and facilitate angiogenic processes that are important for cutaneous tissue regeneration.
Collapse
|
10
|
Hutchings G, Janowicz K, Moncrieff L, Dompe C, Strauss E, Kocherova I, Nawrocki MJ, Kruszyna Ł, Wąsiatycz G, Antosik P, Shibli JA, Mozdziak P, Perek B, Krasiński Z, Kempisty B, Nowicki M. The Proliferation and Differentiation of Adipose-Derived Stem Cells in Neovascularization and Angiogenesis. Int J Mol Sci 2020; 21:ijms21113790. [PMID: 32471255 PMCID: PMC7312564 DOI: 10.3390/ijms21113790] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
Neovascularization and angiogenesis are vital processes in the repair of damaged tissue, creating new blood vessel networks and increasing oxygen and nutrient supply for regeneration. The importance of Adipose-derived Mesenchymal Stem Cells (ASCs) contained in the adipose tissue surrounding blood vessel networks to these processes remains unknown and the exact mechanisms responsible for directing adipogenic cell fate remain to be discovered. As adipose tissue contains a heterogenous population of partially differentiated cells of adipocyte lineage; tissue repair, angiogenesis and neovascularization may be closely linked to the function of ASCs in a complex relationship. This review aims to investigate the link between ASCs and angiogenesis/neovascularization, with references to current studies. The molecular mechanisms of these processes, as well as ASC differentiation and proliferation are described in detail. ASCs may differentiate into endothelial cells during neovascularization; however, recent clinical trials have suggested that ASCs may also stimulate angiogenesis and neovascularization indirectly through the release of paracrine factors.
Collapse
Affiliation(s)
- Greg Hutchings
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Krzysztof Janowicz
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Lisa Moncrieff
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Claudia Dompe
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Correspondence:
| | - Ewa Strauss
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland;
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Ievgeniia Kocherova
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Mariusz J. Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Łukasz Kruszyna
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Grzegorz Wąsiatycz
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
| | - Jamil A. Shibli
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, São Paulo 07023-070, Brazil;
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland;
| | - Zbigniew Krasiński
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| |
Collapse
|
11
|
Liu CH, Huang S, Britton WR, Chen J. MicroRNAs in Vascular Eye Diseases. Int J Mol Sci 2020; 21:ijms21020649. [PMID: 31963809 PMCID: PMC7014392 DOI: 10.3390/ijms21020649] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of the first microRNA (miRNA) decades ago, studies of miRNA biology have expanded in many biomedical research fields, including eye research. The critical roles of miRNAs in normal development and diseases have made miRNAs useful biomarkers or molecular targets for potential therapeutics. In the eye, ocular neovascularization (NV) is a leading cause of blindness in multiple vascular eye diseases. Current anti-angiogenic therapies, such as anti-vascular endothelial growth factor (VEGF) treatment, have their limitations, indicating the need for investigating new targets. Recent studies established the roles of various miRNAs in the regulation of pathological ocular NV, suggesting miRNAs as both biomarkers and therapeutic targets in vascular eye diseases. This review summarizes the biogenesis of miRNAs, and their functions in the normal development and diseases of the eye, with a focus on clinical and experimental retinopathies in both human and animal models. Discovery of novel targets involving miRNAs in vascular eye diseases will provide insights for developing new treatments to counter ocular NV.
Collapse
Affiliation(s)
| | | | | | - Jing Chen
- Correspondence: ; Tel.: +1-617-919-2525
| |
Collapse
|
12
|
Roy S, Glaser S, Chakraborty S. Inflammation and Progression of Cholangiocarcinoma: Role of Angiogenic and Lymphangiogenic Mechanisms. Front Med (Lausanne) 2019; 6:293. [PMID: 31921870 PMCID: PMC6930194 DOI: 10.3389/fmed.2019.00293] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA), or cancer of the biliary epithelium is a relatively rare but aggressive form of biliary duct cancer which has a 5-year survival rate post metastasis of 2%. Although a number of risk factors are established for CCA growth and progression, a careful evaluation of the existing literature on CCA reveals that an inflammatory environment near the biliary tree is the most common causal link between the risk factors and the development of CCA. The fact that inflammation predisposes affected individuals to CCA is further bolstered by multiple observations where the presence and maintenance of an inflammatory microenvironment at the site of the primary tumor plays a significant role in the development and metastasis of CCA. In addition, mechanisms activating the tumor vasculature and enhancing angiogenesis and lymphangiogenesis significantly contribute to CCA aggressiveness and metastasis. This review aims to address the role of an inflammatory microenvironment-CCA crosstalk and will present the basic concepts, observations, and current perspectives from recent research studies in the field of tumor stroma of CCA.
Collapse
Affiliation(s)
- Sukanya Roy
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| |
Collapse
|
13
|
Boen JRA, Gevaert AB, De Keulenaer GW, Van Craenenbroeck EM, Segers VFM. The role of endothelial miRNAs in myocardial biology and disease. J Mol Cell Cardiol 2019; 138:75-87. [PMID: 31756323 DOI: 10.1016/j.yjmcc.2019.11.151] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
The myocardium is a highly structured pluricellular tissue which is governed by an intricate network of intercellular communication. Endothelial cells are the most abundant cell type in the myocardium and exert crucial roles in both healthy myocardium and during myocardial disease. In the last decade, microRNAs have emerged as new actors in the regulation of cellular function in almost every cell type. Here, we review recent evidence on the regulatory function of different microRNAs expressed in endothelial cells, also called endothelial microRNAs, in healthy and diseased myocardium. Endothelial microRNA emerged as modulators of angiogenesis in the myocardium, they are implicated in the paracrine role of endothelial cells in regulating cardiac contractility and homeostasis, and interfere in the crosstalk between endothelial cells and cardiomyocytes.
Collapse
Affiliation(s)
- Jente R A Boen
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Andreas B Gevaert
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, ZNA Middelheim Hospital, Lindendreef 1, 2020 Antwerp, Belgium.
| | - Emeline M Van Craenenbroeck
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Vincent F M Segers
- Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
14
|
Roma-Rodrigues C, Fernandes AR, Baptista PV. Counteracting the effect of leukemia exosomes by antiangiogenic gold nanoparticles. Int J Nanomedicine 2019; 14:6843-6854. [PMID: 31692567 PMCID: PMC6716571 DOI: 10.2147/ijn.s215711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Progression of chronic myeloid leukemia (CML) is frequently associated with increased angiogenesis at the bone marrow mediated by exosomes. The capability of gold nanoparticles (AuNPs) functionalized with antiangiogenic peptides to hinder the formation of new blood vessels has been demonstrated in a chorioallantoic membrane (CAM) model. Methods Exosomes of K562 CML cell line were isolated and their angiogenic effect assessed in a CAM model. AuNPs functionalized with antiangiogenic peptides were used to block the angiogenic effect of CML-derived exosomes, assessed by evaluation of expression levels of key modulators involved in angiogenic pathways - VEGFA, VEGFR1 (also known as FLT1) and IL8. Results Exosomes isolated from K562 cells promoted the doubling of newly formed vessels associated with the increase of VEGFR1 expression. This is a concentration and time-dependent effect. The AuNPs functionalized with antiangiogenic peptides were capable to block the angiogenic effect by modulating VEGFR1 associated pathway. Conclusion Exosomes derived from blast cells are capable to trigger (neo)-angiogenesis, a key factor for the progression and spreading of cancer, in particular in CML. AuNPs functionalized with specific antiangiogenic peptides are capable to block the effect of the exosomes produced by malignant cells via modulation of the intrinsic VEGFR pathway. Together, these data highlight the potential of nanomedicine-based strategies against cancer proliferation.
Collapse
Affiliation(s)
- Catarina Roma-Rodrigues
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica 2829-516, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica 2829-516, Portugal
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica 2829-516, Portugal
| |
Collapse
|
15
|
Barbui T, De Stefano V, Falanga A, Finazzi G, Martinelli I, Rodeghiero F, Vannucchi AM, Barosi G. Addressing and proposing solutions for unmet clinical needs in the management of myeloproliferative neoplasm-associated thrombosis: A consensus-based position paper. Blood Cancer J 2019; 9:61. [PMID: 31395856 PMCID: PMC6687826 DOI: 10.1038/s41408-019-0225-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 12/18/2022] Open
Abstract
This article presents the results of a group discussion among an ad hoc constituted Panel of experts aimed at highlighting unmet clinical needs (UCNs) in the management of thrombotic risk and thrombotic events associated with Philadelphia-negative myeloproliferative neoplasms (Ph-neg MPNs). With the Delphi technique, the challenges in Ph-neg MPN-associated thrombosis were selected. The most clinically relevant UCNs resulted in: (1) providing evidence of the benefits and risks of direct oral anticoagulants, (2) providing evidence of the benefits and risks of cytoreduction in patients with splanchnic vein thrombosis without hypercythemia, (3) improving knowledge of the role of the mutated endothelium in the pathogenesis of thrombosis, (4) improving aspirin dosing regimens in essential thrombocythemia, (5) improving antithrombotic management of Ph-neg MPN-associated pregnancy, (6) providing evidence for the optimal duration of anticoagulation for prophylaxis of recurrent VTE, (7) improving knowledge of the association between somatic gene mutations and risk factors for thrombosis, and (8) improving the grading system of thrombosis risk in polycythemia vera. For each of these issues, proposals for advancement in research and clinical practice were addressed. Hopefully, this comprehensive overview will serve to inform the design and implementation of new studies in the field.
Collapse
Affiliation(s)
- Tiziano Barbui
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy.
| | - Valerio De Stefano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Istituto di Ematologia, Università Cattolica, Roma, Italy
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine and the Haemostasis and Thrombosis Center, Papa Giovanni XXIII Hospital, Bergamo, Italy.,University of Milan Bicocca, Milan, Italy
| | - Guido Finazzi
- Hematology Division, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Ida Martinelli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, A. Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Francesco Rodeghiero
- Hematology Project Foundation, affiliated to the Department of Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Alessandro M Vannucchi
- CRIMM-Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, Dept Experimental and Clinical medicine, and Denothe Center, University of Florence, Florence, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
16
|
Micrornas at the Interface between Osteogenesis and Angiogenesis as Targets for Bone Regeneration. Cells 2019; 8:cells8020121. [PMID: 30717449 PMCID: PMC6406308 DOI: 10.3390/cells8020121] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Bone formation and regeneration is a multistep complex process crucially determined by the formation of blood vessels in the growth plate region. This is preceded by the expression of growth factors, notably the vascular endothelial growth factor (VEGF), secreted by osteogenic cells, as well as the corresponding response of endothelial cells, although the exact mechanisms remain to be clarified. Thereby, coordinated coupling between osteogenesis and angiogenesis is initiated and sustained. The precise interplay of these two fundamental processes is crucial during times of rapid bone growth or fracture repair in adults. Deviations in this balance might lead to pathologic conditions such as osteoarthritis and ectopic bone formation. Besides VEGF, the recently discovered important regulatory and modifying functions of microRNAs also support this key mechanism. These comprise two principal categories of microRNAs that were identified with specific functions in bone formation (osteomiRs) and/or angiogenesis (angiomiRs). However, as hypoxia is a major driving force behind bone angiogenesis, a third group involved in this process is represented by hypoxia-inducible microRNAs (hypoxamiRs). This review was focused on the identification of microRNAs that were found to have an active role in osteogenesis as well as angiogenesis to date that were termed "CouplingmiRs (CPLGmiRs)". Outlined representatives therefore represent microRNAs that already have been associated with an active role in osteogenic-angiogenic coupling or are presumed to have its potential. Elucidation of the molecular mechanisms governing bone angiogenesis are of great relevance for improving therapeutic options in bone regeneration, tissue-engineering, and the treatment of bone-related diseases.
Collapse
|
17
|
Wu JB, Tang YL, Liang XH. Targeting VEGF pathway to normalize the vasculature: an emerging insight in cancer therapy. Onco Targets Ther 2018; 11:6901-6909. [PMID: 30410348 PMCID: PMC6200071 DOI: 10.2147/ott.s172042] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vascular normalization is a new concept of targeting angiogenesis to restore vessel structure and function and to increase blood perfusion and delivery of drugs. It has been confirmed that vascular normalization can decrease relapse and benefit other cancer therapy, including chemotherapy, radiotherapy, and immune cell therapy. The key point of this therapy is to inhibit pro-angiogenic factors and make it be balanced with anti-angiogenic factors, resulting in a mature and normal vessel characteristic. Vascular endothelial growth factor (VEGF) is a key player in the process of tumor angiogenesis, and inhibiting VEGF is a primary approach to tumor vessel normalization. Herein, we review newly uncovered mechanisms governing angiogenesis and vascular normalization of cancer and place emphasis on targeting VEGF pathway to normalize the vasculature. Also, important methods to depress VEGF pathway and make tumor vascular are discussed.
Collapse
Affiliation(s)
- Jing-Biao Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| |
Collapse
|
18
|
Pi J, Liu J, Zhuang T, Zhang L, Sun H, Chen X, Zhao Q, Kuang Y, Peng S, Zhou X, Yu Z, Tao T, Tomlinson B, Chan P, Tian Y, Fan H, Liu Z, Zheng X, Morrisey E, Zhang Y. Elevated Expression of miR302-367 in Endothelial Cells Inhibits Developmental Angiogenesis via CDC42/CCND1 Mediated Signaling Pathways. Theranostics 2018; 8:1511-1526. [PMID: 29556338 PMCID: PMC5858164 DOI: 10.7150/thno.21986] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022] Open
Abstract
Rationale: Angiogenesis is critical for embryonic development and microRNAs fine-tune this process, but the underlying mechanisms remain incompletely understood. Methods: Endothelial cell (EC) specific miR302-367 line was used as gain-of-function and anti-miRs as loss-of-function models to investigate the effects of miR302-367 on developmental angiogenesis with embryonic hindbrain vasculature as an in vivo model and fibrin gel beads and tube formation assay as in vitro models. Cell migration was evaluated by Boyden chamber and scratch wound healing assay and cell proliferation by cell count, MTT assay, Ki67 immunostaining and PI cell cycle analysis. RNA high-throughput sequencing identified miR-target genes confirmed by chromatin immunoprecipitation and 3'-UTR luciferase reporter assay, and finally target site blocker determined the pathway contributing significantly to the phenotype observed upon microRNA expression. Results: Elevated EC miR302-367 expression reduced developmental angiogenesis, whereas it was enhanced by inhibition of miR302-367, possibly due to the intrinsic inhibitory effects on EC migration and proliferation. We identified Cdc42 as a direct target gene and elevated EC miR302-367 decreased total and active Cdc42, and further inhibited F-actin formation via the WASP and Klf2/Grb2/Pak1/LIM-kinase/Cofilin pathways. MiR302-367-mediated-Klf2 regulation of Grb2 for fine-tuning Pak1 activation contributing to the inhibited F-actin formation, and then the attenuation of EC migration. Moreover, miR302-367 directly down-regulated EC Ccnd1 and impaired cell proliferation via the Rb/E2F pathway. Conclusion: miR302-367 regulation of endothelial Cdc42 and Ccnd1 signal pathways for EC migration and proliferation advances our understanding of developmental angiogenesis, and meanwhile provides a rationale for future interventions of pathological angiogenesis that shares many common features of physiological angiogenesis.
Collapse
|
19
|
van Beijnum JR, Giovannetti E, Poel D, Nowak-Sliwinska P, Griffioen AW. miRNAs: micro-managers of anticancer combination therapies. Angiogenesis 2017; 20:269-285. [PMID: 28474282 PMCID: PMC5519663 DOI: 10.1007/s10456-017-9545-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/11/2017] [Indexed: 12/15/2022]
Abstract
Angiogenesis is one of the hallmarks of cancer progression and as such has been considered a target of therapeutic interest. However, single targeted agents have not fully lived up to the initial promise of anti-angiogenic therapy. Therefore, it has been suggested that combining therapies and agents will be the way forward in the oncology field. In recent years, microRNAs (miRNAs) have received considerable attention as drivers of tumor development and progression, either acting as tumor suppressors or as oncogenes (so-called oncomiRs), as well as in the process of tumor angiogenesis (angiomiRs). Not only from a functional, but also from a therapeutic view, miRNAs are attractive tools. Thus far, several mimics and antagonists of miRNAs have entered clinical development. Here, we review the provenance and promise of miRNAs as targets as well as therapeutics to contribute to anti-angiogenesis-based (combination) treatment of cancer.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Laboratory Medical Oncology, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Dennis Poel
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Varinska L, Kubatka P, Mojzis J, Zulli A, Gazdikova K, Zubor P, Büsselberg D, Caprnda M, Opatrilova R, Gasparova I, Klabusay M, Pec M, Fibach E, Adamek M, Kruzliak P. Angiomodulators in cancer therapy: New perspectives. Biomed Pharmacother 2017; 89:578-590. [PMID: 28258040 DOI: 10.1016/j.biopha.2017.02.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/03/2017] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
The formation of new blood vessels plays a crucial for the development and progression of pathophysiological changes associated with a variety of disorders, including carcinogenesis. Angiogenesis inhibitors (anti-angiogenics) are an important part of treatment for some types of cancer. Some natural products isolated from marine invertebrates have revealed antiangiogenic activities, which are diverse in structure and mechanisms of action. Many preclinical studies have generated new models for further modification and optimization of anti-angiogenic substances, and new information for mechanistic studies and new anti-cancer drug candidates for clinical practice. Moreover, in the last decade it has become apparent that galectins are important regulators of tumor angiogenesis, as well as microRNA. MicroRNAs have been validated to modulate endothelial cell migration or endothelial tube organization. In the present review we summarize the current knowledge regarding the role of marine-derived natural products, galectins and microRNAs in tumor angiogenesis.
Collapse
Affiliation(s)
- Lenka Varinska
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia; Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia.
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Anthony Zulli
- The Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Werribee Campus, Victoria, Australia
| | - Katarina Gazdikova
- Department of Nutrition, Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovak Republic; Department of General Medicine, Faculty of Medicine, Slovak Medical University, Bratislava, Slovak Republic.
| | - Pavol Zubor
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia; Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Dietrich Büsselberg
- Weill Cornell Medicine in Qatar, Qatar Foundation-Education City, Doha, Qatar
| | - Martin Caprnda
- 2nd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Radka Opatrilova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho tr. 1/1946, 612 42 Brno, Czechia
| | - Iveta Gasparova
- Institute of Biology, Genetics and Medical Genetics, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovak Republic
| | - Martin Klabusay
- Department of Haemato-Oncology and Department of Internal Medicine - Cardiology, Faculty of Medicine, Palacky University, Olomouc, Czechia
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Eitan Fibach
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Mariusz Adamek
- Department of Thoracic Surgery, Faculty of Medicine and Dentistry, Medical University of Silesia, Katowice, Poland
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho tr. 1/1946, 612 42 Brno, Czechia.
| |
Collapse
|
21
|
Lin HM, Mahon KL, Spielman C, Gurney H, Mallesara G, Stockler MR, Bastick P, Briscoe K, Marx G, Swarbrick A, Horvath LG. Phase 2 study of circulating microRNA biomarkers in castration-resistant prostate cancer. Br J Cancer 2017; 116:1002-1011. [PMID: 28278515 PMCID: PMC5396108 DOI: 10.1038/bjc.2017.50] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 12/19/2022] Open
Abstract
Background: Biomarkers of therapeutic response and prognosis are needed to assist in the sequencing of treatments for metastatic castration-resistant prostate cancer (CRPC). Previously in a Phase 1 discovery study, we identified 14 circulating microRNAs that were associated with response to docetaxel chemotherapy or overall survival. We performed a Phase 2 validation study to verify these findings. Methods: Using real-time PCR, the levels of the 14 microRNAs were measured in plasma collected before and after the first cycle of docetaxel from a Phase 2 cohort of 89 patients. Results: The microRNAs were not associated with docetaxel response in the Phase 2 cohort. Higher baseline levels of six microRNAs, predominantly of the miR-200 family, were confirmed to be associated with shorter overall survival. A microRNA signature comprising these six microRNAs predicted high-risk patients in the Phase 2 cohort with a hazard ratio of 4.12 (95% CI 2.20–7.70, P=0.000001). The signature was an independent predictor in multivariable analysis with clinicopathological factors. Conclusions: The association of circulating microRNAs with overall survival suggests their involvement in CRPC progression.
Collapse
Affiliation(s)
- Hui-Ming Lin
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Darlinghurst, New South Wales 2010, Australia
| | - Kate L Mahon
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Darlinghurst, New South Wales 2010, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia.,Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| | - Calan Spielman
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Howard Gurney
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales 2145, Australia.,Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia
| | - Girish Mallesara
- Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,Calvary Mater Newcastle Hospital, Waratah, New South Wales 2298, Australia
| | - Martin R Stockler
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia.,Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia.,Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia.,Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia
| | - Patricia Bastick
- Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,St George Hospital, Kogarah, New South Wales 2217, Australia
| | - Karen Briscoe
- Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,Mid North Coast Cancer Institute, Coffs Harbour Health Campus, Coffs Harbour, New South Wales 2450, Australia
| | - Gavin Marx
- Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,Sydney Adventist Hospital, Wahroonga, New South Wales 2076, Australia
| | - Alexander Swarbrick
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Darlinghurst, New South Wales 2010, Australia
| | - Lisa G Horvath
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Darlinghurst, New South Wales 2010, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia.,Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia.,Pharmacogenomics Research for Individualised Medicine (PRIMe) consortium, New South Wales, Australia.,Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia
| |
Collapse
|
22
|
Rawal S, Munasinghe PE, Shindikar A, Paulin J, Cameron V, Manning P, Williams MJA, Jones GT, Bunton R, Galvin I, Katare R. Down-regulation of proangiogenic microRNA-126 and microRNA-132 are early modulators of diabetic cardiac microangiopathy. Cardiovasc Res 2017; 113:90-101. [PMID: 28065883 DOI: 10.1093/cvr/cvw235] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 07/15/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM Microangiopathy due to endothelial dysfunction is a major contributing factor to the development of diabetes-induced cardiovascular disease (CVD). Dysregulation of endothelial-specific microRNAs (miRs) is correlated with impaired angiogenesis and cell survival. We investigated the profile of two angiomiRs, miR-126, and miR-132, in the plasma of type 2 diabetic individuals without any known history of CVD as well as in the cardiac tissues collected from diabetics undergoing cardiac surgery. METHODS AND RESULTS The presence of diabetes alone significantly decreased both angiomiRs in the plasma and the myocardium. The down-regulation of angiomiRs was also associated with reduced capillaries and arterioles and increased endothelial cell apoptosis, the hallmark of microangiopathy. Importantly, a time course study in a type 2 diabetic mouse model confirmed that the down-regulation of angiomiRs preceded endothelial apoptosis as well as alterations in the density of the microvasculature. Finally, therapeutic overexpression of both angiomiRs in diabetic aortic rings and human umbilical vein endothelial cells exposed to high glucose (HG) abrogated the deleterious effects of diabetes and HG on cell survival and proliferation and restored their angiogenic potential. CONCLUSIONS These novel findings demonstrate that the down-regulation of angiomiRs is a major underlying mechanism for the development of microangiopathy in diabetic hearts. Therefore, therapeutic restoration of angiomiRs could become a potential approach to combat the cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | | | - Amol Shindikar
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | - Jono Paulin
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | | | | | | | | | - Richard Bunton
- Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago
| | - Ivor Galvin
- Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago
| | - Rajesh Katare
- Department of Physiology-HeartOtago, Otago School of Medical Sciences;
| |
Collapse
|
23
|
Generation of functional endothelial cells with progenitor-like features from murine induced pluripotent stem cells. Vascul Pharmacol 2016; 86:94-108. [DOI: 10.1016/j.vph.2016.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/20/2016] [Indexed: 11/19/2022]
|
24
|
Pi J, Tao T, Zhuang T, Sun H, Chen X, Liu J, Cheng Y, Yu Z, Zhu HH, Gao WQ, Suo Y, Wei X, Chan P, Zheng X, Tian Y, Morrisey E, Zhang L, Zhang Y. A MicroRNA302-367-Erk1/2-Klf2-S1pr1 Pathway Prevents Tumor Growth via Restricting Angiogenesis and Improving Vascular Stability. Circ Res 2016; 120:85-98. [PMID: 27756792 DOI: 10.1161/circresaha.116.309757] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/02/2016] [Accepted: 10/18/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE Angiogenic hypersprouting and leaky vessels are essential for tumor growth. MicroRNAs have unique therapeutic advantages by targeting multiple pathways of tumor-associated angiogenesis, but the function of individual miRNAs of miR302-367 cluster in angiogenesis and tumors has not yet been fully evaluated. OBJECTIVE To investigate the functions of miR302-367 in developmental angiogenesis and tumor angiogenesis and explore the molecular mechanisms of microRNA for the treatment of pathological neovascularization-related diseases. METHODS AND RESULTS Here, we show that miR302-367 elevation in endothelial cells reduces retinal sprouting angiogenesis and promotes vascular stability in vivo, ex vivo, and in vitro. Erk1/2 is identified as direct target of miR302-367, and downregulation of Erk1/2 on miR302-367 elevation in endothelial cells increases the expression of Klf2 and in turn S1pr1 and its downstream target VE-cadherin, suppressing angiogenesis and improving vascular stability. Conversely, both pharmacological blockade and genetic deletion of S1pr1 in endothelial cells reverse the antiangiogenic and vascular stabilizing effect of miR302-367 in mice. Tumor angiogenesis shares features of developmental angiogenesis, and endothelial specific elevation of miR302-367 reduces tumor growth by restricting sprout angiogenesis and decreasing vascular permeability via the same Erk1/2-Klf2-S1pr1 pathways. CONCLUSIONS MiR302-367 regulation of an Erk1/2-Klf2-S1pr1 pathway in the endothelium advances our understanding of angiogenesis, meanwhile also provides opportunities for therapeutic intervention of tumor growth.
Collapse
Affiliation(s)
- Jingjiang Pi
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Ting Tao
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Tao Zhuang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Huimin Sun
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Xiaoli Chen
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Jie Liu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Yu Cheng
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Zuoren Yu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Helen He Zhu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Wei-Qiang Gao
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Yuanzhen Suo
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Xunbin Wei
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Paul Chan
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Xiangjian Zheng
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Ying Tian
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Edward Morrisey
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Lin Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - YuZhen Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (J.P., T.Z., H.S., X.C., J.L., Z.Y., L.Z., Y.Z.); Department of Geriatrics, Ruijin Hospital, School of Medicine (T.T.) and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, (Y.S., X.W.), Shanghai Jiao Tong University, China; Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China (Y.C.); Med-X-Renji Hospital Stem Cell Research Center, Jiao Tong University School of Medicine, Shanghai, China (H.H.Z., W.-Q.G.); Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.); Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, New South Wales, Australia (X.Z.); Department of Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia (X.Z.); Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (Y.T.); and Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.).
| |
Collapse
|
25
|
Jour G, Ivan D, Aung PP. Angiogenesis in melanoma: an update with a focus on current targeted therapies. J Clin Pathol 2016; 69:472-83. [PMID: 26865640 DOI: 10.1136/jclinpath-2015-203482] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/02/2016] [Indexed: 12/29/2022]
Abstract
Angiogenesis plays a crucial role in melanoma metastasis and progression. In recent years, numerous studies have investigated the prognostic and clinical significance of this phenomenon, and the development of molecular techniques has enabled us to achieve a better understanding of angiogenesis in melanoma. Herein, we review the current state of knowledge regarding angiogenesis in melanoma, including the pathophysiological, histological and immunohistochemical aspects of this phenomenon. We also review the molecular pathways involved in angiogenesis and the interplay between different components that might be manipulated in the future development of efficient targeted therapies. Recently developed targeted antiangiogenic therapies in clinical trials and included in the treatment of advanced-stage melanoma are also reviewed.
Collapse
Affiliation(s)
- George Jour
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Doina Ivan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Avallone A, Piccirillo MC, Aloj L, Nasti G, Delrio P, Izzo F, Di Gennaro E, Tatangelo F, Granata V, Cavalcanti E, Maiolino P, Bianco F, Aprea P, De Bellis M, Pecori B, Rosati G, Carlomagno C, Bertolini A, Gallo C, Romano C, Leone A, Caracò C, de Lutio di Castelguidone E, Daniele G, Catalano O, Botti G, Petrillo A, Romano GM, Iaffaioli VR, Lastoria S, Perrone F, Budillon A. A randomized phase 3 study on the optimization of the combination of bevacizumab with FOLFOX/OXXEL in the treatment of patients with metastatic colorectal cancer-OBELICS (Optimization of BEvacizumab scheduLIng within Chemotherapy Scheme). BMC Cancer 2016; 16:69. [PMID: 26857924 PMCID: PMC4746902 DOI: 10.1186/s12885-016-2102-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the improvements in diagnosis and treatment, colorectal cancer (CRC) is the second cause of cancer deaths in both sexes. Therefore, research in this field remains of great interest. The approval of bevacizumab, a humanized anti-vascular endothelial growth factor (VEGF) monoclonal antibody, in combination with a fluoropyrimidine-based chemotherapy in the treatment of metastatic CRC has changed the oncology practice in this disease. However, the efficacy of bevacizumab-based treatment, has thus far been rather modest. Efforts are ongoing to understand the better way to combine bevacizumab and chemotherapy, and to identify valid predictive biomarkers of benefit to avoid unnecessary and costly therapy to nonresponder patients. The BRANCH study in high-risk locally advanced rectal cancer patients showed that varying bevacizumab schedule may impact on the feasibility and efficacy of chemo-radiotherapy. METHODS/DESIGN OBELICS is a multicentre, open-label, randomised phase 3 trial comparing in mCRC patients two treatment arms (1:1): standard concomitant administration of bevacizumab with chemotherapy (mFOLFOX/OXXEL regimen) vs experimental sequential bevacizumab given 4 days before chemotherapy, as first or second treatment line. Primary end point is the objective response rate (ORR) measured according to RECIST criteria. A sample size of 230 patients was calculated allowing reliable assessment in all plausible first-second line case-mix conditions, with a 80% statistical power and 2-sided alpha error of 0.05. Secondary endpoints are progression free-survival (PFS), overall survival (OS), toxicity and quality of life. The evaluation of the potential predictive role of several circulating biomarkers (circulating endothelial cells and progenitors, VEGF and VEGF-R SNPs, cytokines, microRNAs, free circulating DNA) as well as the value of the early [(18)F]-Fluorodeoxyglucose positron emission tomography (FDG-PET) response, are the objectives of the traslational project. DISCUSSION Overall this study could optimize bevacizumab scheduling in combination with chemotherapy in mCRC patients. Moreover, correlative studies could improve the knowledge of the mechanisms by which bevacizumab enhance chemotherapy effect and could identify early predictors of response. EudraCT Number: 2011-004997-27 TRIAL REGISTRATION: ClinicalTrials.gove number, NCT01718873.
Collapse
Affiliation(s)
- Antonio Avallone
- Multidisciplinary Treatment Unit, Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy. .,Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | | | - Luigi Aloj
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Guglielmo Nasti
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Paolo Delrio
- Colorectal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Bianco
- Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Pasquale Aprea
- Vascular Access Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Mario De Bellis
- Endoscopy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Biagio Pecori
- Radiotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Gerardo Rosati
- Medical Oncology Unit, San Carlo Hospital, Potenza, Italy.
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.
| | | | - Ciro Gallo
- Medical Statistics Unit, Second University, Naples, Italy.
| | - Carmela Romano
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Corradina Caracò
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | | | - Gennaro Daniele
- Clinical Trials Unit, Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Napoli, Italy.
| | - Orlando Catalano
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Giovanni M Romano
- Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Vincenzo R Iaffaioli
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Napoli, Italy.
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| |
Collapse
|
27
|
Abstract
Solid tumors require angiogenesis to grow beyond 2 mm in size. In most cases, tumor cells undergo angiogenic switch and secrete substances that are required for generation of new capillary sprouting from existing blood vessels. Tumor angiogenesis is driven by a complex interplay between pro-angiogenic (VEGF/VEGFR, PDGF/PDGFR) and anti-angiogenic factors (TSP-1/TSP-2) within the tumor microenvironment. In addition, control of tissue remodeling and degradation by matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) contribute to tumor angiogenesis. Furthermore, tumor suppressors or oncogenes that control cellular motility and maintain or promote hypoxia (HIFs and MYC) are also actively playing roles in tumor angiogenesis. Noncoding RNAs (ncRNAs), including microRNAs, are a novel class of regulatory molecules that control the gene expression in a posttranscriptional manner. MicroRNAs regulate important physiological processes, such as proliferation, apoptosis, and differentiation, as well as pathological conditions including oncogenesis. Accumulating evidence suggests that microRNAs directly modulate the process of angiogenesis by targeting important angiogenic factors and signaling molecules. Understanding the molecular mechanism behind the regulation of angiogenesis by microRNAs is important due to their therapeutic potential which may lead to improving outcome for cancer patients. Besides, ncRNAs with a regulatory role in angiogenesis, such as long noncoding RNAs (lncRNAs), have been identified in the genome. However, the mechanisms of the vast majority of lncRNAs are currently unknown. For the few lncRNAs characterized at the functional level, accumulating evidence shows that they play important roles in malignant diseases. The function and mechanism in angiogenesis will be described in this chapter.
Collapse
|
28
|
Distinct microRNA expression in endometrial lymphocytes, endometrium, and trophoblast during spontaneous porcine fetal loss. J Reprod Immunol 2014; 107:64-79. [PMID: 25596873 DOI: 10.1016/j.jri.2014.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/18/2014] [Accepted: 11/24/2014] [Indexed: 01/22/2023]
Abstract
Endometrial lymphocytes are recruited to the porcine maternal-fetal interface by conceptus-derived signals. The transiently recruited lymphocytes adopt a specialized phenotype in the endometrium that regulates various placental physiological processes, including angiogenesis. Small non-coding RNAs, microRNAs (miRNAs) are emerging as principal bio-molecules regulating the development of lymphocytes and their angiogenic functions. However, no information is available in the context of endometrial lymphocytes in pregnancy. We hypothesize that miRNAs are involved in the development of endometrial lymphocytes and their angiogenic functions at the porcine maternal-fetal interface. Using a targeted Q-PCR approach for selected miRNAs involved in immune cell development, angiogenesis, and anti-angiogenesis, we conducted a study to screen endometrial lymphocytes associated with healthy and spontaneously arresting conceptus attachment sites (CAS) at two well-defined periods of fetal loss. Comparisons were made with endometrium and trophoblasts associated with healthy and arresting CAS. In addition, levels of putative mRNA targets and subsequent functional clustering of genes were studied in order to predict the biological mechanisms affected. We found several significant differences for miRNAs involved in immune cell development and angiogenesis (miR-296-5P, miR-150, miR-17P-5P, miR-18a, and miR-19a) between endometrial lymphocytes associated with healthy and arresting CAS. Significant differences were also found in endometrium and trophoblasts for some miRNAs (miR-20b, miR-17-5P, miR-18a, miR-15b-5P, and miR-222). Finally, selected mRNA targets showed differential expression in all groups. Our data, although associative, are the first to unravel the selected miRNAs involved in immune cell development and provide insights into their possible regulation in abortive pregnancy.
Collapse
|
29
|
Lønvik K, Sørbye SW, Nilsen MN, Paulssen RH. Prognostic value of the MicroRNA regulators Dicer and Drosha in non-small-cell lung cancer: co-expression of Drosha and miR-126 predicts poor survival. BMC Clin Pathol 2014; 14:45. [PMID: 25525410 PMCID: PMC4269969 DOI: 10.1186/1472-6890-14-45] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/03/2014] [Indexed: 12/26/2022] Open
Abstract
Background Dicer and Drosha are important enzymes for processing microRNAs. Recent studies have exhibited possible links between expression of different miRNAs, levels of miRNA processing enzymes, and cancer prognosis. We have investigated the prognostic impact of Dicer and Drosha and their correlation with miR-126 expression in a large cohort of non-small cell lung cancer (NSCLC) patients. We aimed to find patient groups within the cohort that might have an advantage of receiving adjunctive therapies. Methods Dicer expression in the cytoplasm and Drosha expression in the nucleus were evaluated by manual immunohistochemistry of tissue microarrays (TMAs), including tumor tissue samples from 335 patients with resected stages I to IIIA NSCLC. In addition, in situ hybridizations of TMAs for visualization of miR-126 were performed. Kaplan–Meier analysis was performed, and the log-rank test via SPSS v.22 was used for estimating significance levels. Results In patients with normal performance status (ECOG = 0, n = 197), high Dicer expression entailed a significantly better prognosis than low Dicer expression (P = 0.024). Dicer had no significant prognostic value in patients with reduced performance status (ECOG = 1–2, n = 138). High Drosha expression was significantly correlated with high levels of the microRNA 126 (miR-126) (P = 0.004). Drosha/miR-126 co-expression had a significant negative impact on the disease-specific survival (DSS) rate (P < 0.001). Multivariate analyses revealed that the interaction Dicer*Histology (P = 0.049) and Drosha/miR-126 co-expression (P = 0.033) were independent prognostic factors. Conclusions In NSCLC patients with normal performance status, Dicer is a positive prognostic factor. The importance of Drosha as a prognostic factor in our material seems to be related to miR-126 and possibly other microRNAs. Electronic supplementary material The online version of this article (doi:10.1186/1472-6890-14-45) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kenneth Lønvik
- Department of Clinical Pathology, University Hospital of Northern Norway, N-9038 Tromsø, Norway ; Department of Medical Biology, Tromsø, Norway
| | - Sveinung W Sørbye
- Department of Clinical Pathology, University Hospital of Northern Norway, N-9038 Tromsø, Norway
| | | | - Ruth H Paulssen
- Department of Clinical Medicine, UiT - The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
30
|
Squadrito ML, De Palma M. Micromanaging restenosis by therapeutic inhibition of miR-92a. Cardiovasc Res 2014; 103:432-4. [PMID: 25082845 DOI: 10.1093/cvr/cvu178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Mario Leonardo Squadrito
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
31
|
Xu X, Kriegel AJ, Jiao X, Liu H, Bai X, Olson J, Liang M, Ding X. miR-21 in ischemia/reperfusion injury: a double-edged sword? Physiol Genomics 2014; 46:789-97. [PMID: 25159851 DOI: 10.1152/physiolgenomics.00020.2014] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are endogenous, small RNA molecules that suppress expression of targeted mRNA. miR-21, one of the most extensively studied miRNAs, is importantly involved in divergent pathophysiological processes relating to ischemia/reperfusion (I/R) injury, such as inflammation and angiogenesis. The role of miR-21 in renal I/R is complex, with both protective and pathological pathways being regulated by miR-21. Preconditioning-induced upregulation of miR-21 contributes to the protection against subsequent renal I/R injury through the targeting of genes such as the proapoptotic gene programmed cell death 4 and interactions between miR-21 and hypoxia-inducible factor. Conversely, long-term elevation of miR-21 may be detrimental to the organ by promoting the development of renal interstitial fibrosis following I/R injury. miR-21 is importantly involved in several pathophysiological processes related to I/R injury including inflammation and angiogenesis as well as the biology of stem cells that could be used to treat I/R injury; however, the effect of miR-21 on these processes in renal I/R injury remains to be studied.
Collapse
Affiliation(s)
- Xialian Xu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoyan Jiao
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Hong Liu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jessica Olson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoqiang Ding
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China; Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, Peoples Republic of China; Kidney and Dialysis Institute of Shanghai, Shanghai, Peoples Republic of China; and Kidney and Blood Purification Laboratory of Shanghai, Shanghai, Peoples Republic of China
| |
Collapse
|
32
|
Agrawal S, Chaqour B. MicroRNA signature and function in retinal neovascularization. World J Biol Chem 2014; 5:1-11. [PMID: 24600510 PMCID: PMC3942538 DOI: 10.4331/wjbc.v5.i1.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/11/2013] [Accepted: 01/06/2014] [Indexed: 02/05/2023] Open
Abstract
Ischemic retinopathies are clinically well-defined chronic microvascular complications characterized by gradually progressive alterations in the retinal microvasculature and a compensatory aberrant neovascularization of the eye. The subsequent metabolic deficiencies result in structural and functional alterations in the retina which is highly susceptible to injurious stimuli such as diabetes, trauma, hyperoxia, inflammation, aging and dysplipidemia. Emerging evidence indicates that an effective therapy may require targeting multiple components of the angiogenic pathway. Conceptually, mircoRNA (miRNA)-based therapy provides the rationale basis for an effective antiangiogenic treatment. miRNAs are an evolutionarily conserved family of short RNAs, each regulating the expression of multiple protein-coding genes. The activity of specific miRNAs is important for vascular cell signaling and blood vessel formation and function. Recently, important progress has been made in mapping the miRNA-gene target network and miRNA-mediated gene expression control. Here we highlight the latest findings on angiogenic and antiangiogenic miRNAs and their targets as well as potential implications in ocular neovascular diseases. Emphasis is placed on how specific vascular-enriched miRNAs regulate cell responses to various cues by targeting several factors, receptors and/or signaling molecules in order to maintain either vascular function or dysfunction. Further improvement of our knowledge in not only miRNA specificity, turnover, and transport but also how miRNA sequences and functions can be altered will enhance the therapeutic utility of such molecules.
Collapse
|
33
|
Rawal S, Manning P, Katare R. Cardiovascular microRNAs: as modulators and diagnostic biomarkers of diabetic heart disease. Cardiovasc Diabetol 2014; 13:44. [PMID: 24528626 PMCID: PMC3976030 DOI: 10.1186/1475-2840-13-44] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/10/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetic heart disease (DHD) is the leading cause of morbidity and mortality among the people with diabetes, with approximately 80% of the deaths in diabetics are due to cardiovascular complications. Importantly, heart disease in the diabetics develop at a much earlier stage, although remaining asymptomatic till the later stage of the disease, thereby restricting its early detection and active therapeutic management. Thus, a better understanding of the modulators involved in the pathophysiology of DHD is necessary for the early diagnosis and development of novel therapeutic implications for diabetes-associated cardiovascular complications. microRNAs (miRs) have recently been evolved as key players in the various cardiovascular events through the regulation of cardiac gene expression. Besides their credible involvement in controlling the cellular processes, they are also released in to the circulation in disease states where they serve as potential diagnostic biomarkers for cardiovascular disease. However, their potential role in DHD as modulators as well as diagnostic biomarkers is largely unexplored. In this review, we describe the putative mechanisms of the selected cardiovascular miRs in relation to cardiovascular diseases and discuss their possible involvement in the pathophysiology and early diagnosis of DHD.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Physiology, HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Patrick Manning
- Department of Medicine, Dunedin Hospital, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology, HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
34
|
Liles WC, Kain KC. Endothelial activation and dysfunction in the pathogenesis of microvascular obstruction in severe malaria--a viable target for therapeutic adjunctive intervention. J Infect Dis 2014; 210:163-4. [PMID: 24443542 DOI: 10.1093/infdis/jiu035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
35
|
Shi L, Fisslthaler B, Zippel N, Frömel T, Hu J, Elgheznawy A, Heide H, Popp R, Fleming I. MicroRNA-223 Antagonizes Angiogenesis by Targeting β1 Integrin and Preventing Growth Factor Signaling in Endothelial Cells. Circ Res 2013; 113:1320-30. [DOI: 10.1161/circresaha.113.301824] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rationale:
Endothelial cells in situ are largely quiescent, and their isolation and culture are associated with the switch to a proliferative phenotype.
Objective:
To identify antiangiogenic microRNAs expressed by native endothelial cells that are altered after isolation and culture, as well as the protein targets that regulate responses to growth factors.
Methods and Results:
Profiling studies revealed that miR-223 was highly expressed in freshly isolated human, murine, and porcine endothelial cells, but those levels decreased in culture. In primary cultures of endothelial cells, vascular endothelial cell growth factor and basic fibroblast growth factor further decreased miR-223 expression. The overexpression of precursor-miR-223 did not affect basal endothelial cell proliferation but abrogated vascular endothelial cell growth factor–induced and basic fibroblast growth factor–induced proliferation, as well as migration and sprouting. Inhibition of miR-223 in vivo using specific antagomirs potentiated postnatal retinal angiogenesis in wild-type mice, whereas recovery of perfusion after femoral artery ligation and endothelial sprouting from aortic rings from adult miR-223
−/y
animals were enhanced. MiR-223 overexpression had no effect on the growth factor–induced activation of ERK1/2 but inhibited the vascular endothelial cell growth factor–induced and basic fibroblast growth factor–induced phosphorylation of their receptors and activation of Akt. β1 integrin was identified as a target of miR-223 and its downregulation reproduced the defects in growth factor receptor phosphorylation and Akt signaling seen after miR-223 overexpression. Reintroduction of β1 integrin into miR-223–ovexpressing cells was sufficient to rescue growth factor signaling and angiogenesis.
Conclusions:
These results indicate that miR-223 is an antiangiogenic microRNA that prevents endothelial cell proliferation at least partly by targeting β1 integrin.
Collapse
Affiliation(s)
- Lei Shi
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Beate Fisslthaler
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Nina Zippel
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Timo Frömel
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Jiong Hu
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Amro Elgheznawy
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Heinrich Heide
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Rüdiger Popp
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| | - Ingrid Fleming
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, Frankfurt, Germany (L.S., B.F., N.Z., T.F., J.H., A.E., R.P., I.F.); and Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany (H.H.)
| |
Collapse
|
36
|
Devalliere J, Chang WG, Andrejecsk JW, Abrahimi P, Cheng CJ, Jane-wit D, Saltzman WM, Pober JS. Sustained delivery of proangiogenic microRNA-132 by nanoparticle transfection improves endothelial cell transplantation. FASEB J 2013; 28:908-22. [PMID: 24221087 DOI: 10.1096/fj.13-238527] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Transplantation of endothelial cells (ECs) for therapeutic vascularization or tissue engineering is a promising method for increasing tissue perfusion. Here, we report on a new approach for enhanced EC transplantation using targeted nanoparticle transfection to deliver proangiogenic microRNA-132 (miR-132) to cultured ECs before their transplantation, thereby sensitizing cells to the effects of endogenous growth factors. We synthesized biodegradable PLGA polymer nanoparticles (NPs) that were loaded with miR-132 and coated with cyclic RGD (cRGD) peptides that target integrin αvβ3 expressed on cultured human umbilical vein ECs (HUVECs), increasing NP uptake through clathrin-coated pits. Unlike previously reported NPs for miR delivery, these NPs slowly release RNA for several weeks. The endocytosed NPs remain in clathrin-coated vesicles from which they mediate intracellular delivery of siRNA or miRNA. Transfection of HUVECs with miR-132 enhances growth factor-induced proliferation and migration in 2D culture, producing a 1.8- and 5-fold increase, respectively. However, while the effects of conventional transfection were short-lived, NP transfection produced protein knockdown and biological effects that were significantly longer in duration (≥ 6 d). Transfection of HUVECs with miR-132 NP resulted in a 2-fold increase in the number of microvessels per square millimeter compared to lipid after transplantation into immunodeficient mice and led to a higher number of mural cell-invested vessels than control transfection. These data suggest that sustained delivery of miR-132 encapsulated in a targeted biodegradable polymer NP is a safe and efficient strategy to improve EC transplantation and vascularization.
Collapse
Affiliation(s)
- Julie Devalliere
- 2Department of Immunobiology, PO Box 208089, New Haven, CT 06520-8089, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Bitzer M, Ben-Dov IZ, Thum T. Microparticles and microRNAs of endothelial progenitor cells ameliorate acute kidney injury. Kidney Int 2013; 82:375-7. [PMID: 22846811 PMCID: PMC3410558 DOI: 10.1038/ki.2012.152] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Horizontal information transfer between cells via microparticles is a newly identified communication system. MicroRNAs regulate gene expression and are detected in microparticles. The article by Cantaluppi et al. suggests that microparticles derived from circulating angiogenic cells (“endothelial progenitor cells”, EPC) harbor endothelial-protective miRNAs such as miR-126 and that delivery of EPC-derived microparticles during acute kidney ischemia-reperfusion in rats ameliorates kidney dysfunction and damage. We highlight the importance, future impact and limitations of this study.
Collapse
|
38
|
Crosstalk-induced loss of miR-126 promotes angiogenesis. Oncogene 2013; 33:3634-5. [PMID: 24013227 DOI: 10.1038/onc.2013.317] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 07/09/2013] [Indexed: 12/13/2022]
Abstract
A molecular environment that promotes vascularization around human carcinomas can materialise rapidly, and has been termed the angiogenic switch. Turning this switch toward a proangiogenic state involves an altered interplay between tumor cells and multiple components of the surrounding stroma. The regulatory landscape of these interactions in cervical cancer is now investigated by Huang et al. in this issue of Oncogene, who demonstrate that the microRNA miR-126 is downregulated during cancer progression, particularly in stromal cells. Such a reduction of miR-126 is shown to free at least one target, the proangiogenic adrenomedullin, from repression, enhancing vascular growth especially at the in situ to invasive carcinoma transition. The study implicates the temporal, spatial and progressive nature of tumor-stroma interactions during carcinogenesis, while in turn suggesting therapeutic strategies.
Collapse
|
39
|
Zheng Z, Zeng Y, Huang H, Xu F. MicroRNA-132 may play a role in coexistence of depression and cardiovascular disease: a hypothesis. Med Sci Monit 2013; 19:438-43. [PMID: 23748239 PMCID: PMC3678976 DOI: 10.12659/msm.883935] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Different individuals have different degrees of neuroplasticity due to their different experiences. Neuroplasticity may play a role in individual differences among neuropsychiatric disease treatment efficacy. Since the nervous system monitors and coordinates internal organ function, neuroplasticity may be associated with other diseases. Cardiovascular disease (CVD) is associated with depression, which is a disorder of disrupted neuroplasticity. MicroRNA-132 (miR-132) has a roles in neuroplasticity and cardiovascular function. Thus, we hypothesize that miR-132 may play a role in coexistence of depression and CVD.
Collapse
Affiliation(s)
- Zhihua Zheng
- Guangdong Province Pharmaceutical Association, Guangzhou, China.
| | | | | | | |
Collapse
|
40
|
Wang RY, Phang RZ, Hsu PH, Wang WH, Huang HT, Liu IY. In vivo knockdown of hippocampal miR-132 expression impairs memory acquisition of trace fear conditioning. Hippocampus 2013; 23:625-33. [PMID: 23520022 DOI: 10.1002/hipo.22123] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/31/2022]
Abstract
MicroRNA-132 (miR-132) has been demonstrated to affect multiple neuronal functions, including dendritic growth and spinogenesis in cultured neurons and brain slices, as well as learning behavior of animals. However, its role in acquisition of temporal-associated memory remains unclear. In this study, we demonstrated that the mature miR-132 level in mouse hippocampus was significantly increased at 30 min after trace fear conditioning, a type of temporal-associated learning, and returned to baseline values in 2 h. We then knocked down miR-132 expression in vivo by infusing a lentivector expressing anti-miR-132 hairpin RNA into the third ventricle near the anterior hippocampi such RNA diffused laterally to both hippocampal formations, later confirmed by histological analysis. This approach successfully reduced hippocampal miR-132 expression in both naïve and trace fear conditioned groups, and impaired acquisition of trace fear memory in mice. To our knowledge, this result is the first demonstration of change in temporal learning behavior by reducing microRNA (miRNA) level specifically in the hippocampal region.
Collapse
Affiliation(s)
- Ruo-Yu Wang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, 970, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Loss of glomerular function associated with the presence of tubulointerstitial lesions, which are characterized by peritubular capillary loss, is a common finding in progressive renal disorders. Dysregulated expression of angiogenic factors (such as vascular endothelial growth factor [VEGF] and angiopoietins) and endogenous angiogenic inhibitors (such as thrombospondin-1, angiostatin and endostatin) underlie these conditions and negatively influence the balance between capillary formation and regression, resulting in capillary rarefaction. Recent studies have provided unequivocal evidence for a pathogenic role of tubulointerstitial hypoxia and the involvement of hypoxia-inducible transcription factors in the advanced stages of chronic kidney disease. The mainstay of potential angiogenic therapies is the application of angiogenic factors with the primary aim of ameliorating reduced oxygenation in the ischaemic tubulointerstitium. However, this strategy is strongly associated with inflammation and changes in vascular permeability. For example, supraphysiological expression of VEGF results in glomerular expansion and proteinuria, whereas VEGF blockade using neutralizing antibodies can cause hypertension and thrombotic microangiopathy. These effects highlight the importance of tight regulation of angiogenic factors and inhibitors. Novel therapeutic approaches that target vascular maturation and normalization are now being developed to protect kidneys from capillary rarefaction and hypoxic injury.
Collapse
Affiliation(s)
- Tetsuhiro Tanaka
- Division for Health Service Promotion, University of Tokyo School of Medicine, Tokyo, Japan
| | | |
Collapse
|
42
|
Ando H, Okamoto A, Yokota M, Shimizu K, Asai T, Dewa T, Oku N. Development of a miR-92a delivery system for anti-angiogenesis-based cancer therapy. J Gene Med 2013; 15:20-7. [DOI: 10.1002/jgm.2690] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/02/2012] [Accepted: 12/10/2012] [Indexed: 01/31/2023] Open
Affiliation(s)
- Hidenori Ando
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| | - Ayaka Okamoto
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| | - Masafumi Yokota
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| | - Kosuke Shimizu
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| | - Takehisa Dewa
- Department of Life and Materials Engineering; Nagoya Institute of Technology; Nagoya; Japan
| | - Naoto Oku
- Department of Medical Biochemistry; University of Shizuoka Graduate School of Pharmaceutical Sciences; Shizuoka; Japan
| |
Collapse
|
43
|
Guduric-Fuchs J, O'Connor A, Cullen A, Harwood L, Medina RJ, O'Neill CL, Stitt AW, Curtis TM, Simpson DA. Deep sequencing reveals predominant expression of miR-21 amongst the small non-coding RNAs in retinal microvascular endothelial cells. J Cell Biochem 2012; 113:2098-111. [PMID: 22298343 PMCID: PMC3708110 DOI: 10.1002/jcb.24084] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The retinal vascular endothelium is essential for angiogenesis and is involved in maintaining barrier selectivity and vascular tone. The aim of this study was to identify and quantify microRNAs and other small regulatory non-coding RNAs (ncRNAs) which may regulate these crucial functions. Primary bovine retinal microvascular endothelial cells (RMECs) provide a well-characterized in vitro system for studying angiogenesis. RNA extracted from RMECs was used to prepare a small RNA library for deep sequencing (Illumina Genome Analyzer). A total of 6.8 million reads were mapped to 250 known microRNAs in miRBase (release 16). In many cases, the most frequent isomiR differed from the sequence reported in miRBase. In addition, five novel microRNAs, 13 novel bovine orthologs of known human microRNAs and multiple new members of the miR-2284/2285 family were detected. Several ∼30 nucleotide sno-miRNAs were identified, with the most highly expressed being derived from snoRNA U78. Highly expressed microRNAs previously associated with endothelial cells included miR-126 and miR-378, but the most highly expressed was miR-21, comprising more than one-third of all mapped reads. Inhibition of miR-21 with an LNA inhibitor significantly reduced proliferation, migration, and tube-forming capacity of RMECs. The independence from prior sequence knowledge provided by deep sequencing facilitates analysis of novel microRNAs and other small RNAs. This approach also enables quantitative evaluation of microRNA expression, which has highlighted the predominance of a small number of microRNAs in RMECs. Knockdown of miR-21 suggests a role for this microRNA in regulation of angiogenesis in the retinal microvasculature. J. Cell. Biochem. 113: 2098–2111, 2012. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jasenka Guduric-Fuchs
- Centre for Vision and Vascular Science, Queen's University Belfast, Belfast, Northern Ireland, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Recent findings indicate that specific microRNAs (miRNAs), such as those of the miR-17-92 cluster, may be responsible for regulating endothelial gene expression during tumor angiogenesis. Secreted miRNAs enclosed in exosomes also have an important role in cell-cell communication. To elucidate whether miRNAs secreted from neoplastic cells transfer into endothelial cells and are functionally active in the recipient cells, we investigated the effect of exosomal miRNAs derived from leukemia cells (K562) on human umbilical vein endothelial cells (HUVECs). As K562 cells released the miR-17-92 cluster, especially miR-92a, into the extracellular environment, K562 cells, transfected with Cy3-labeled pre-miR-92a, were co-cultured with HUVECs. Cy3-miR-92a derived from K562 cells was detected in the cytoplasm of HUVECs, and the Cy3-miR-92a co-localized with the signals of an exosomal marker, CD63. The expression of integrin α5, a target gene for miR-92a, was significantly reduced in HUVECs by exosomal miR-92a, indicating that exogenous miRNA via exosomal transport can function like endogenous miRNA in HUVECs. The most salient feature of this study is the exosome, derived from K562 cells with enforced miR-92a expression, did not affect the growth of HUVECs but did enhance endothelial cell migration and tube formation. Our results support the idea that exosomal miRNAs have an important role in neoplasia-to-endothelial cell communication.
Collapse
|
45
|
Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J 2012; 31:3513-23. [PMID: 22773185 DOI: 10.1038/emboj.2012.183] [Citation(s) in RCA: 386] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 06/14/2012] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis plays a crucial role during tumorigenesis and much progress has been recently made in elucidating the role of VEGF and other growth factors in the regulation of angiogenesis. Recently, microRNAs (miRNAs) have been shown to modulate a variety of physiogical and pathological processes. We identified a set of differentially expressed miRNAs in microvascular endothelial cells co-cultured with tumour cells. Unexpectedly, most miRNAs were derived from tumour cells, packaged into microvesicles (MVs), and then directly delivered to endothelial cells. Among these miRNAs, we focused on miR-9 due to the strong morphological changes induced in cultured endothelial cells. We found that exogenous miR-9 effectively reduced SOCS5 levels, leading to activated JAK-STAT pathway. This signalling cascade promoted endothelial cell migration and tumour angiogenesis. Remarkably, administration of anti-miR-9 or JAK inhibitors suppressed MV-induced cell migration in vitro and decreased tumour burden in vivo. Collectively, these observations suggest that tumour-secreted miRNAs participate in intercellular communication and function as a novel pro-angiogenic mechanism.
Collapse
|
46
|
Mendias CL, Gumucio JP, Lynch EB. Mechanical loading and TGF-β change the expression of multiple miRNAs in tendon fibroblasts. J Appl Physiol (1985) 2012; 113:56-62. [PMID: 22539168 DOI: 10.1152/japplphysiol.00301.2012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tendons link skeletal muscles to bones and are important components of the musculoskeletal system. There has been much interest in the role of microRNA (miRNA) in the regulation of musculoskeletal tissues to mechanical loading, inactivity, and disease, but it was unknown whether miRNA is involved in the adaptation of tendons to mechanical loading. We hypothesized that mechanical loading and transforming growth factor-β (TGF-β) treatment would regulate the expression of several miRNA molecules with known roles in cell proliferation and extracellular matrix synthesis. To test our hypothesis, we subjected untrained adult rats to a single session of mechanical loading and measured the expression of several miRNA transcripts in Achilles tendons. Additionally, as TGF-β is known to be an important regulator of tendon growth and adaptation, we treated primary tendon fibroblasts with TGF-β and measured miRNA expression. Both mechanical loading and TGF-β treatment modulated the expression of several miRNAs that regulate cell proliferation and extracellular matrix synthesis. We also identified mechanosensitive miRNAs that may bind to the 3'-untranslated region of the basic helix-loop-helix transcription factor scleraxis, which is a master regulator of limb tendon development. The results from this study provide novel insight into the mechanobiology of tendons and indicate that miRNA could play an important role in the adaptation of tendons to growth stimuli.
Collapse
Affiliation(s)
- Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.
| | | | | |
Collapse
|
47
|
De Palma M, Hanahan D. The biology of personalized cancer medicine: facing individual complexities underlying hallmark capabilities. Mol Oncol 2012; 6:111-27. [PMID: 22360993 PMCID: PMC5528366 DOI: 10.1016/j.molonc.2012.01.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 01/29/2012] [Indexed: 12/14/2022] Open
Abstract
It is a time of great promise and expectation for the applications of knowledge about mechanisms of cancer toward more effective and enduring therapies for human disease. Conceptualizations such as the hallmarks of cancer are providing an organizing principle with which to distill and rationalize the abject complexities of cancer phenotypes and genotypes across the spectrum of the human disease. A countervailing reality, however, involves the variable and often transitory responses to most mechanism-based targeted therapies, returning full circle to the complexity, arguing that the unique biology and genetics of a patient's tumor will in the future necessarily need to be incorporated into the decisions about optimal treatment strategies, the frontier of personalized cancer medicine. This perspective highlights considerations, metrics, and methods that may prove instrumental in charting the landscape of evaluating individual tumors so to better inform diagnosis, prognosis, and therapy. Integral to the consideration is remarkable heterogeneity and variability, evidently embedded in cancer cells, but likely also in the cell types composing the supportive and interactive stroma of the tumor microenvironment (e.g., leukocytes and fibroblasts), whose diversity in form, regulation, function, and abundance may prove to rival that of the cancer cells themselves. By comprehensively interrogating both parenchyma and stroma of patients' cancers with a suite of parametric tools, the promise of mechanism-based therapy may truly be realized.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Douglas Hanahan
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
48
|
Collet G, Skrzypek K, Grillon C, Matejuk A, El Hafni-Rahbi B, Lamerant-Fayel N, Kieda C. Hypoxia control to normalize pathologic angiogenesis: potential role for endothelial precursor cells and miRNAs regulation. Vascul Pharmacol 2012; 56:252-61. [PMID: 22446152 DOI: 10.1016/j.vph.2012.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 02/18/2012] [Accepted: 03/02/2012] [Indexed: 01/12/2023]
Abstract
Tumor microenvironment is a complex and highly dynamic milieu that provides very important clues on tumor development and progression mechanisms. Tumor-associated endothelial cells play a key role in stroma organization. They achieve tumor angiogenesis, a formation of tumor-associated (angiogenic) vessels mainly through sprouting from locally preexisting vessels and/or recruitment of bone marrow-derived endothelial progenitor cells. This process participates to supply nutritional support and oxygen to the growing tumor. Endothelial cells constitute the interface between circulating blood cells, tumor cells and the extracellular matrix, thereby controlling leukocyte recruitment, tumor cell behavior and metastasis formation. Hypoxia, a critical parameter of the tumor microenvironment, controls endothelial/tumor cell interactions and is the key to tumor angiogenesis development. Under hypoxic stress, tumor cells produce factors that promote angiogenesis, vasculogenesis, tumor cell motility, metastasis and cancer stem cell selection. Targeting tumor vessels is a therapeutic strategy that has lately been fast evolving from antiangiogenesis to vessel normalization as discussed in this review. We shall focus on the pivotal role of endothelial cells within the tumor microenvironment, the specific features and the part played by circulating endothelial precursors cells. Attention is stressed on their recruitment to the tumor site and their role in tumor angiogenesis where they are submitted to miRNAs-mediated de/regulation. Here the compensation of the tumor deregulated angiogenic miRNAs - angiomiRs - is emphasized as a potential therapeutic approach. The strategy is to over express anti-angiomiRs in the tumor angiogenesis site upon selective delivery by precursor endothelial cells as miRs carriers.
Collapse
Affiliation(s)
- Guillaume Collet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, 45071 Orleans, France
| | | | | | | | | | | | | |
Collapse
|
49
|
Fitzpatrick LE, Chan JW, Sefton MV. On the mechanism of poly(methacrylic acid –co– methyl methacrylate)-induced angiogenesis: Gene expression analysis of dTHP-1 cells. Biomaterials 2011; 32:8957-67. [DOI: 10.1016/j.biomaterials.2011.08.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/09/2011] [Indexed: 01/13/2023]
|
50
|
Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 2011; 17:1359-70. [PMID: 22064426 DOI: 10.1038/nm.2537] [Citation(s) in RCA: 1305] [Impact Index Per Article: 93.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|