151
|
Newman J, Brailovsky Y, Allen S, Bontekoe E, Masic D, Walenga J, Fareed J, Darki A. Angiopoietin-2 correlates with pulmonary embolism severity, right ventricular dysfunction, and intensive care unit admission. Vasc Med 2021; 26:556-560. [PMID: 33840325 DOI: 10.1177/1358863x211002276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Risk stratification of acute pulmonary embolism (PE) is important to identify patients at risk for hemodynamic collapse who would benefit from more aggressive therapies. Angiopoietin-2 (Ang-2) is a signaling molecule involved in angiogenesis and is upregulated in response to tissue hypoxia. We aimed to assess the association of Ang-2 with (1) PE severity, (2) echocardiographic and invasive hemodynamic markers of right ventricular (RV) dysfunction, and (3) need for intensive treatment. Patients presenting to our institution with acute PE were included in a prospective database and blood samples were collected and stored for later analysis. A total of 65 patients were included in the study. Ang-2 correlated with PE risk stratification and echocardiographic and invasive hemodynamic markers of RV dysfunction and pulmonary hypertension. An Ang-2 level of > 4101 pg/mL had an odds ratio of 7.4 (95% CI: 1.53-12.5, p < 0.01) for intensive care unit (ICU) admission. In conclusion, Ang-2 correlates with PE severity, RV dysfunction, and need for ICU admission. Ang-2 holds promise as a novel marker that can aid in risk stratification for this patient population.
Collapse
Affiliation(s)
- Joshua Newman
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Yevgeniy Brailovsky
- Advanced Heart Failure, Mechanical Circulatory Support, Heart Transplantation, Jefferson Heart Institute, Sidney Kimmel School of Medicine, Philadelphia, PA, USA
| | - Sorcha Allen
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Emily Bontekoe
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Dalila Masic
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Jeanine Walenga
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Jawed Fareed
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Amir Darki
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
152
|
Pinto-Bravo P, Rebordão MR, Amaral A, Fernandes C, Galvão A, Silva E, Pessa-Santos P, Alexandre-Pires G, Roberto da Costa RP, Skarzynski DJ, Ferreira-Dias G. Microvascularization and Expression of Fibroblast Growth Factor and Vascular Endothelial Growth Factor and Their Receptors in the Mare Oviduct. Animals (Basel) 2021; 11:ani11041099. [PMID: 33921416 PMCID: PMC8070128 DOI: 10.3390/ani11041099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The oviduct provides the ideal conditions for fertilization and early embryonic development. Adequate vascularization is essential for proper oviduct physiological function. In this work on the mare oviduct, differences in the oviductal artery and arterioles and their ramifications in the infundibulum, ampulla and isthmus were examined. Locally, vascularization is modulated by the action of angiogenic factors, mediated by their specific receptors. In the present study, the isthmus presented the largest vascular area and the highest number of vascular structures in the follicular phase. We have also shown that the relative abundance of angiogenic transcripts and proteins, such as fibroblast growth factor 1 (FGF1) and 2 (FGF2) and vascular endothelial growth factor (VEGF), and their respective receptors (FGFR1, FGFR2, VEGFR2 = KDR), were present in all portions of the oviduct throughout the estrous cycle. There was an increase in the transcripts of angiogenic receptors FGF1 and FGFR1 in the ampulla and isthmus, and of FGF2 and KDR in the isthmus. This was also observed in the isthmus, where the relative abundance of proteins FGFR1 and KDR was the highest. This study shows that the equine oviduct presents differences in microvascular density in its portions. The angiogenic factors VEGF, FGF1, FGF2 and their respective receptors are expressed in all studied regions of the mare oviduct, in agreement with microvascular patterns. Abstract The oviduct presents the ideal conditions for fertilization and early embryonic development. In this study, (i) vascularization pattern; (ii) microvascular density; (iii) transcripts of angiogenic factors (FGF1, FGF2, VEGF) and their receptors—FGFR1, FGFR2, KDR, respectively, and (iv) the relative protein abundance of those receptors were assessed in cyclic mares’ oviducts. The oviductal artery, arterioles and their ramifications, viewed by means of vascular injection-corrosion, differed in the infundibulum, ampulla and isthmus. The isthmus, immunostained with CD31, presented the largest vascular area and the highest number of vascular structures in the follicular phase. Transcripts (qPCR) and relative protein abundance (Western blot) of angiogenic factors fibroblast growth factor 1 (FGF1) and 2 (FGF2) and vascular endothelial growth factor (VEGF), and their respective receptors (FGFR1, FGFR2, VEGFR2 = KDR), were present in all oviduct portions throughout the estrous cycle. Upregulation of the transcripts of angiogenic receptors FGF1 and FGFR1 in the ampulla and isthmus and of FGF2 and KDR in the isthmus were noted. Furthermore, in the isthmus, the relative protein abundance of FGFR1 and KDR was the highest. This study shows that the equine oviduct presents differences in microvascular density in its three portions. The angiogenic factors VEGF, FGF1, FGF2 and their respective receptors are expressed in all studied regions of the mare oviduct, in agreement with microvascular patterns.
Collapse
Affiliation(s)
- Pedro Pinto-Bravo
- CERNAS (Research Center for Natural Resources, Environment and Society), Polytechnic Institute of Coimbra, 3045-601 Coimbra, Portugal; (P.P.-B.); (R.P.R.d.C.)
- Coimbra College of Agriculture, Polytechnic Institute of Coimbra, 3045-601 Coimbra, Portugal;
| | - Maria Rosa Rebordão
- Coimbra College of Agriculture, Polytechnic Institute of Coimbra, 3045-601 Coimbra, Portugal;
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
| | - Ana Amaral
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
| | - Carina Fernandes
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
| | - António Galvão
- Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10-748 Olsztyn, Poland; (A.G.); (D.J.S.)
| | - Elisabete Silva
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
| | | | - Graça Alexandre-Pires
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
| | - Rosário P. Roberto da Costa
- CERNAS (Research Center for Natural Resources, Environment and Society), Polytechnic Institute of Coimbra, 3045-601 Coimbra, Portugal; (P.P.-B.); (R.P.R.d.C.)
- Coimbra College of Agriculture, Polytechnic Institute of Coimbra, 3045-601 Coimbra, Portugal;
| | - Dariusz J. Skarzynski
- Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10-748 Olsztyn, Poland; (A.G.); (D.J.S.)
| | - Graça Ferreira-Dias
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (A.A.); (C.F.); (E.S.); (G.A.-P.)
- Correspondence: ; Tel.: +351-213-652-859
| |
Collapse
|
153
|
Abdel Ghafar MT, Elkhouly RA, Elnaggar MH, Mabrouk MM, Darwish SA, Younis RL, Elkholy RA. Utility of serum neuropilin-1 and angiopoietin-2 as markers of hepatocellular carcinoma. J Investig Med 2021; 69:1222-1229. [PMID: 33833047 DOI: 10.1136/jim-2020-001744] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/24/2022]
Abstract
This study aimed to assess the diagnostic value of two serum angiogenetic markers neuropilin-1 (NRP-1) and angiopoietin-2 (ANG-2) in patients with hepatocellular carcinoma (HCC) and their relation to tumor characteristics. 149 subjects were recruited and divided into 50 patients with recently diagnosed HCC, 49 patients with cirrhosis on top of hepatitis C virus infection, and 50 healthy subjects. Serum NRP-1 and ANG-2 were estimated by ELISA. Alpha-fetoprotein (AFP) levels were measured using fluorescence immunoassay. Serum NRP-1 and ANG-2 levels were significantly higher in patients with HCC (2221.8±1056.6 pg/mL and 3018.5±841.4 pg/mL) than healthy subjects (219.3±61.8 pg/mL and 2007.7±904.8 pg/mL) and patients with cirrhosis (1108.9±526.6 pg/mL and 2179.1±599.2 pg/mL), respectively. In multivariate logistic regression analysis, NRP-1 and AFP were the only independent factors of HCC development and correlated positively with each other (r=0.781, p<0.001). Receiver operating characteristic curve analysis showed that the area under the curve (AUC) of NRP-1 was higher than that of ANG-2 in discriminating HCC from patients with cirrhosis (0.801 vs 0.748, p=0.250) and healthy subjects (0.992 vs 0.809, p<0.001). The AUC of NRP-1 was detected to be increased (0.994) when combined estimation with AFP was performed. Elevated serum NRP-1 and ANG-2 levels were detected in patients with HCC with tumor numbers >3, tumor size ≥5 cm, tumor stages B/C according to the Barcelona Clinic Liver Cancer staging system, vascular invasion, and distant metastasis. In conclusion, NRP-1 is a potential serological marker for HCC diagnosis and is better than ANG-2. It is feasible to be estimated in combination with AFP to enhance its diagnostic power. High serum NRP-1 and ANG-2 levels are associated with advanced HCC tumor characteristics.
Collapse
Affiliation(s)
| | - Reham A Elkhouly
- Tropical Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | | | - Mohamed M Mabrouk
- Internal Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Sara A Darwish
- Clinical Oncology and Nuclear Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Reham L Younis
- Physiology, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Rasha A Elkholy
- Clinical Pathology, Tanta University Faculty of Medicine, Tanta, Egypt
| |
Collapse
|
154
|
Man RJ, Jeelani N, Zhou C, Yang YS. Recent Progress in the Development of Quinoline Derivatives for the Exploitation of Anti-Cancer Agents. Anticancer Agents Med Chem 2021; 21:825-838. [PMID: 32416703 DOI: 10.2174/1871520620666200516150345] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/23/2020] [Accepted: 02/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Along with the progress in medicine and therapies, the exploitation of anti-cancer agents focused more on the vital signaling pathways and key biological macromolecules. With rational design and advanced synthesis, quinoline derivatives have been utilized frequently in medicinal chemistry, especially in developing anti-cancer drugs or candidates. METHODS Using DOI searching, articles published before 2020 all over the world have been reviewed as comprehensively as possible. RESULTS In this review, we selected the representative quinoline derivate drugs in market or clinical trials, classified them into five major categories with detailed targets according to their main mechanisms, discussed the relationship within the same mechanism, and generated a summative discussion with prospective expectations. For each mechanism, the introduction of the target was presented, with the typical examples of quinoline derivate drugs. CONCLUSION This review has highlighted the quinoline drugs or candidates, suited them into corresponding targets in their pathways, summarized and discussed. We hope that this review may help the researchers who are interested in discovering quinoline derivate anti-cancer agents obtain considerable understanding of this specific topic. Through the flourishing period and the vigorous strategies in clinical trials, quinoline drugs would be potential but facing new challenges in the future.
Collapse
Affiliation(s)
- Ruo-Jun Man
- College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China
| | - Nasreen Jeelani
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Chongchen Zhou
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Yu-Shun Yang
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
155
|
Rajamanickam A, Munisankar S, Menon PA, Nutman TB, Babu S. Diminished circulating levels of angiogenic factors and RAGE ligands in helminth-diabetes comorbidity and reversal following anthelmintic treatment. J Infect Dis 2021; 224:1614-1622. [PMID: 33822083 DOI: 10.1093/infdis/jiab170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Various epidemiological and experimental studies propose that helminths could play a preventive role against the progression of Type 2 diabetes mellitus (T2DM). T2DM induces microvascular and large vessel complications mediated by elevated levels of angiogenic factors and soluble RAGE ligands. However, the interactions between helminths and host angiogenic factors and RAGE ligands are unexplored. METHODS To assess the relationship between a soil-transmitted helminth, Strongyloides stercoralis (Ss) and T2DM, we measured plasma levels of VEGF-A, C, D, Angio-1 and Angio-2 and their receptors VEGF-R1, R2 and R3 as well as sRAGE and their ligands AGE, S100A12 and HMBG-1 in individuals with T2DM with Ss+ or without Ss infection (Ss-). In Ss+ individuals, we also measured the levels of aforementioned factors 6 months following anthelmintic therapy. RESULTS Ss+ individuals exhibited significantly decreased levels of VEGF-A, C, D, Angio-1 and Angio-2 and their soluble receptors VEGF-R1, R2 and R3, that increased following anthelmintic therapy. Likewise, Ss+ individuals exhibited significantly decreased levels of AGE and their ligands sRAGE, S100A12 and HMBG-1 which reversed following anthelmintic therapy. CONCLUSION Our data suggest that Ss infection could play a beneficial role by limiting or delaying the T2DM related vascular complications.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | - Saravanan Munisankar
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | - Pradeep A Menon
- National Institute for Research in Tuberculosis, Chennai, India
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Subash Babu
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India and Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
156
|
Abnormal bone marrow microenvironment: the “harbor” of acute lymphoblastic leukemia cells. BLOOD SCIENCE 2021; 3:29-34. [PMID: 35402834 PMCID: PMC8975096 DOI: 10.1097/bs9.0000000000000071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Bone marrow (BM) microenvironment regulates and supports the production of blood cells which are necessary to maintain homeostasis. In analogy to normal hematopoiesis, leukemogenesis is originated from leukemic stem cells (LSCs) which gives rise to more differentiated malignant cells. Leukemia cells occupy BM niches and reconstruct them to support leukemogenesis. The abnormal BM niches are the main sanctuary of LSCs where they can evade chemotherapy-induced death and acquire drug resistance. In this review, we focus on the protective effects of BM niche cells on acute lymphoblastic leukemia cells.
Collapse
|
157
|
Dieters-Castator D, Dantonio PM, Piaseczny M, Zhang G, Liu J, Kuljanin M, Sherman S, Jewer M, Quesnel K, Kang EY, Köbel M, Siegers GM, Leask A, Hess D, Lajoie G, Postovit LM. Embryonic protein NODAL regulates the breast tumor microenvironment by reprogramming cancer-derived secretomes. Neoplasia 2021; 23:375-390. [PMID: 33784590 PMCID: PMC8041663 DOI: 10.1016/j.neo.2021.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) is an important mediator of breast cancer progression. Cancer-associated fibroblasts constitute a major component of the TME and may originate from tissue-associated fibroblasts or infiltrating mesenchymal stromal cells (MSCs). The mechanisms by which cancer cells activate fibroblasts and recruit MSCs to the TME are largely unknown, but likely include deposition of a pro-tumorigenic secretome. The secreted embryonic protein NODAL is clinically associated with breast cancer stage and promotes tumor growth, metastasis, and vascularization. Herein, we show that NODAL expression correlates with the presence of activated fibroblasts in human triple-negative breast cancers and that it directly induces Cancer-associated fibroblasts phenotypes. We further show that NODAL reprograms cancer cell secretomes by simultaneously altering levels of chemokines (e.g., CXCL1), cytokines (e.g., IL-6) and growth factors (e.g., PDGFRA), leading to alterations in MSC chemotaxis. We therefore demonstrate a hitherto unappreciated mechanism underlying the dynamic regulation of the TME.
Collapse
Affiliation(s)
| | - Paola M Dantonio
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Matt Piaseczny
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Guihua Zhang
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jiahui Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Miljan Kuljanin
- Robarts Research Institute, London, ON, Canada; Department of Biochemistry, Western University, London, ON, Canada
| | - Stephen Sherman
- Robarts Research Institute, London, ON, Canada; Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Michael Jewer
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Katherine Quesnel
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Eun Young Kang
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Andrew Leask
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - David Hess
- Robarts Research Institute, London, ON, Canada; Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Gilles Lajoie
- Department of Biochemistry, Western University, London, ON, Canada
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
158
|
Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Elicit Better Preservation of the Intra-Renal Microvasculature Than Renal Revascularization in Pigs with Renovascular Disease. Cells 2021; 10:cells10040763. [PMID: 33807246 PMCID: PMC8103265 DOI: 10.3390/cells10040763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Percutaneous transluminal renal angioplasty (PTRA) confers clinical and mortality benefits in select ‘high-risk’ patients with renovascular disease (RVD). Intra-renal-delivered extracellular vesicles (EVs) released from mesenchymal stem/stromal cells (MSCs) protect the kidney in experimental RVD, but have not been compared side-by-side to clinically applied interventions, such as PTRA. We hypothesized that MSC-derived EVs can comparably protect the post-stenotic kidney via direct tissue effects. Methods: Five groups of pigs (n = 6 each) were studied after 16 weeks of RVD, RVD treated 4 weeks earlier with either PTRA or MSC-derived EVs, and normal controls. Single-kidney renal blood flow (RBF) and glomerular filtration rate (GFR) were assessed in vivo with multi-detector CT, and renal microvascular architecture (3D micro CT) and injury pathways ex vivo. Results: Despite sustained hypertension, EVs conferred greater improvement of intra-renal microvascular and peritubular capillary density compared to PTRA, associated with attenuation of renal inflammation, oxidative stress, and tubulo-interstitial fibrosis. Nevertheless, stenotic kidney RBF and GFR similarly rose in both PTRA- and EV-treated pigs compared RVD + Sham. mRNA sequencing reveled that EVs were enriched with pro-angiogenic, anti-inflammatory, and antioxidants genes. Conclusion: MSC-derived EVs elicit a better preservation of the stenotic kidney microvasculature and greater attenuation of renal injury and fibrosis compared to PTRA, possibly partly attributed to their cargo of vasculo-protective genes. Yet, both strategies similarly improve renal hemodynamics and function. These observations shed light on diverse mechanisms implicated in improvement of post-stenotic kidney function and position EVs as a promising therapeutic intervention in RVD.
Collapse
|
159
|
Jiao Z, Ma Y, Zhang Q, Wang Y, Liu T, Liu X, Piao C, Liu B, Wang H. The adipose-derived mesenchymal stem cell secretome promotes hepatic regeneration in miniature pigs after liver ischaemia-reperfusion combined with partial resection. Stem Cell Res Ther 2021; 12:218. [PMID: 33781342 PMCID: PMC8008619 DOI: 10.1186/s13287-021-02284-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Background Hepatic ischaemia-reperfusion injury (HIRI) is inevitable in complicated liver surgery and is a major factor leading to postoperative complications and liver dysfunction. Studies have shown that the paracrine mechanisms of stem cell may be essential to tissue repair and functional improvement after transplantation. However, the role of the adipose-derived mesenchymal stem cell secretome (ASC-secretome) in liver regeneration in large animals remains to be determined. Methods Twenty-four miniature pigs were subjected to laparoscopic liver ischaemia-reperfusion combined with partial hepatectomy and divided into the following four groups: the saline group, the DMEM group, the ASC group and the ASC-secretome group. Serum and liver tissue samples were collected before the operation and at 1, 3 and 7 days after the operation, and changes in tissue pathology, serum inflammation, liver function, angiogenesis-related factors and liver tissue regeneration-related genes and proteins were evaluated. Results Detailed histological analysis showed that ASCs and the ASC-secretome changed pathological damage to liver tissue after liver ischaemia-reperfusion combined with partial hepatectomy (1 and 3 days: p < 0.01). Compared with the saline and DMEM control groups, the ASC-secretome group had significantly reduced expression levels of ALP (1 and 3 days: p < 0.05), ALT (1 day: p < 0.01; 3 days: p < 0.05) and AST (1 and 3 days: p < 0.01), which promoted the recovery of liver function. Moreover, detection of the expression levels of TNF-α and IL-1β (1 day: p < 0.01; 3 days: p < 0.05), IL-6 (1 and 3 days: p < 0.05) and IL-10 (1 and 3 days: p < 0.01) in serum confirmed that the ASC-secretome had obvious anti-inflammatory effects. In addition, the ASC-secretome increased the expression levels of ANG-1 (3 days: p < 0.01), ANG-2 (3 and 7 days: p < 0.01) and VEGF (1 and 7 days: p < 0.05; 3 days: p < 0.01) and promoted angiogenesis during liver regeneration. Moreover, it promoted the mRNA expression of HGF and Cyclin D1 (1 and 3 days: p < 0.01); increased the levels of p-STAT3 (1 and 3 days: p < 0.01), PCNA and Ki67 (1 and 3 days: p < 0.01; 7 days: p < 0.05); inhibited the negative feedback of SOCS3 (1 and 3 days: p < 0.01); and decreased the mRNA expression of TGF-β (3 days: p < 0.01). The cytokines and growth factors detected in the ASC-secretome included TNF-α, IL-6, IL-1β, ANG-1, ANG-2, VEGF and b-FGF. Conclusion The ASC-secretome alleviates the inflammatory response induced by ischaemia-reperfusion combined with partial hepatectomy in miniature pigs and promotes liver regeneration.
Collapse
Affiliation(s)
- Zhihui Jiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.,College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qianzhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaoning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Boyang Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.,College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
160
|
Dorschel KB, Wanebo JE. Genetic and Proteomic Contributions to the Pathophysiology of Moyamoya Angiopathy and Related Vascular Diseases. Appl Clin Genet 2021; 14:145-171. [PMID: 33776470 PMCID: PMC7987310 DOI: 10.2147/tacg.s252736] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 12/26/2020] [Indexed: 12/13/2022] Open
Abstract
RATIONALE This literature review describes the pathophysiological mechanisms of the current classes of proteins, cells, genes, and signaling pathways relevant to moyamoya angiopathy (MA), along with future research directions and implementation of current knowledge in clinical practice. OBJECTIVE This article is intended for physicians diagnosing, treating, and researching MA. METHODS AND RESULTS References were identified using a PubMed/Medline systematic computerized search of the medical literature from January 1, 1957, through August 4, 2020, conducted by the authors, using the key words and various combinations of the key words "moyamoya disease," "moyamoya syndrome," "biomarker," "proteome," "genetics," "stroke," "angiogenesis," "cerebral arteriopathy," "pathophysiology," and "etiology." Relevant articles and supplemental basic science articles published in English were included. Intimal hyperplasia, medial thinning, irregular elastic lamina, and creation of moyamoya vessels are the end pathologies of many distinct molecular and genetic processes. Currently, 8 primary classes of proteins are implicated in the pathophysiology of MA: gene-mutation products, enzymes, growth factors, transcription factors, adhesion molecules, inflammatory/coagulation peptides, immune-related factors, and novel biomarker candidate proteins. We anticipate that this article will need to be updated in 5 years. CONCLUSION It is increasingly apparent that MA encompasses a variety of distinct pathophysiologic conditions. Continued research into biomarkers, genetics, and signaling pathways associated with MA will improve and refine our understanding of moyamoya's complex pathophysiology. Future efforts will benefit from multicenter studies, family-based analyses, comparative trials, and close collaboration between the clinical setting and laboratory research.
Collapse
Affiliation(s)
- Kirsten B Dorschel
- Heidelberg University Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - John E Wanebo
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Neuroscience, HonorHealth Research Institute, Scottsdale, AZ, USA
| |
Collapse
|
161
|
Parmar D, Apte M. Angiopoietin inhibitors: A review on targeting tumor angiogenesis. Eur J Pharmacol 2021; 899:174021. [PMID: 33741382 DOI: 10.1016/j.ejphar.2021.174021] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is the process of formation of new blood vessels from existing ones. Vessels serve the purpose of providing oxygen, nutrients and removal of waste from the cells. The physiological angiogenesis is a normal process and is required in the embryonic development, wound healing, menstrual cycle. For homeostasis, balance of pro angiogenic factors and anti angiogenic factors like is important. Their imbalance causes a process known as "angiogenic switch" which leads to various pathological conditions like inflammation, tumor and restenosis. Like normal cells, tumor cells also require oxygen and nutrients to grow which is provided by tumor angiogenesis. Hence angiogenic process can be inhibited to prevent tumor growth. This gives rise to study of anti angiogenic drugs. Currently approved anti angiogenic drugs are mostly VEGF inhibitors, but VEGF inhibitors have certain limitations like toxicity, low progression free survival (PFS), and resistance to anti VEGF therapy. This article focuses on angiopoietins as alternative and potential targets for anti angiogenic therapy. Angiopoietins are ligands of Tie receptor and play a crucial role in angiogenesis, their inhibition can prevent many tumor growths even on later stages of development. We present current clinical and preclinical stages of angiopoietin inhibitors. Drugs studied in the article are selective as well as non-selective inhibitors of angiopoietin 2 like Trebananib (AMG 386), AMG 780, REGN 910, CVX 060, MEDI 3617 and dual inhibitors of angiopoietin 2 and VEGF like Vanucizumab and RG7716. The angiopoietin inhibitors show promising results alone and in combination with VEGF inhibitors in various malignancies.
Collapse
Affiliation(s)
- Digna Parmar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| | - Madhavi Apte
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| |
Collapse
|
162
|
Mosteo L, Storer J, Batta K, Searle EJ, Duarte D, Wiseman DH. The Dynamic Interface Between the Bone Marrow Vascular Niche and Hematopoietic Stem Cells in Myeloid Malignancy. Front Cell Dev Biol 2021; 9:635189. [PMID: 33777944 PMCID: PMC7991089 DOI: 10.3389/fcell.2021.635189] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells interact with bone marrow niches, including highly specialized blood vessels. Recent studies have revealed the phenotypic and functional heterogeneity of bone marrow endothelial cells. This has facilitated the analysis of the vascular microenvironment in steady state and malignant hematopoiesis. In this review, we provide an overview of the bone marrow microenvironment, focusing on refined analyses of the marrow vascular compartment performed in mouse studies. We also discuss the emerging role of the vascular niche in “inflamm-aging” and clonal hematopoiesis, and how the endothelial microenvironment influences, supports and interacts with hematopoietic cells in acute myeloid leukemia and myelodysplastic syndromes, as exemplar states of malignant myelopoiesis. Finally, we provide an overview of strategies for modulating these bidirectional interactions to therapeutic effect in myeloid malignancies.
Collapse
Affiliation(s)
- Laura Mosteo
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joanna Storer
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kiran Batta
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Emma J Searle
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.,Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal.,Department of Onco-Hematology, Instituto Português de Oncologia (IPO)-Porto, Porto, Portugal
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
163
|
Jiang S, Fu R, Shi J, Wu H, Mai J, Hua X, Chen H, Liu J, Lu M, Li N. CircRNA-Mediated Regulation of Angiogenesis: A New Chapter in Cancer Biology. Front Oncol 2021; 11:553706. [PMID: 33777729 PMCID: PMC7988083 DOI: 10.3389/fonc.2021.553706] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is necessary for carcinoma progression and is regulated by a variety of pro- and anti-angiogenesis factors. CircRNAs are RNA molecules that do not have a 5'-cap or a 3'-polyA tail and are involved in a variety of biological functions. While circRNA-mediated regulation of tumor angiogenesis has received much attention, the detailed biological regulatory mechanism remains unclear. In this review, we investigated circRNAs in tumor angiogenesis from multiple perspectives, including its upstream and downstream factors. We believe that circRNAs have natural advantages and great potential for the diagnosis and treatment of tumors, which deserves further exploration.
Collapse
Affiliation(s)
- Shaotao Jiang
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Rongdang Fu
- Department of Hepatic Surgery, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jiewei Shi
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijie Wu
- Department of Obstetrics, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jialuo Mai
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xuefeng Hua
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huan Chen
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie Liu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Minqiang Lu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Li
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
164
|
Yoodee S, Peerapen P, Plumworasawat S, Thongboonkerd V. ARID1A knockdown in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity. Int J Biol Macromol 2021; 180:1-13. [PMID: 33675830 DOI: 10.1016/j.ijbiomac.2021.02.218] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/10/2021] [Accepted: 02/24/2021] [Indexed: 12/29/2022]
Abstract
AT-rich interactive domain 1A (ARID1A) is a novel tumor suppressor gene found in several human cells and its loss/defect is commonly observed in many cancers. However, its roles in angiogenesis, which is one of the hallmarks for tumor progression, remained unclear. Herein, we demonstrated the direct effects of ARID1A knockdown in human endothelial cells by lentivirus-based short-hairpin RNA (shRNA) (shARID1A) on angiogenesis. Functional assays revealed that shARID1A significantly enhanced cell proliferation and migration/invasion and endothelial tube formation compared with the control cells transfected with scramble shRNA (shControl). Additionally, the shARID1A-transfected cells had significantly increased podosome formation and secretion of angiopoietin-2 (ANG2), a key angiogenic factor. Moreover, neutralization of ANG2 with monoclonal anti-ANG2 antibody strongly reduced cell proliferation and migration/invasion and endothelial tube formation in the shARID1A-transfected cells. These findings indicate that down-regulation of ARID1A in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity.
Collapse
Affiliation(s)
- Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Paleerath Peerapen
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
165
|
Angiogenesis in the Normal Adrenal Fetal Cortex and Adrenocortical Tumors. Cancers (Basel) 2021; 13:cancers13051030. [PMID: 33804534 PMCID: PMC7957756 DOI: 10.3390/cancers13051030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/10/2023] Open
Abstract
Angiogenesis plays an important role in several physiological and pathological processes. Pharmacological angiogenesis modulation has been robustly demonstrated to achieve clinical benefits in several cancers. Adrenocortical carcinomas (ACC) are rare tumors that often have a poor prognosis. In addition, therapeutic options for ACC are limited. Understanding the mechanisms that regulate adrenocortical angiogenesis along the embryonic development and in ACC could provide important clues on how these processes could be pharmacologically modulated for ACC treatment. In this report, we performed an integrative review on adrenal cortex angiogenesis regulation in physiological conditions and ACC. During embryonic development, adrenal angiogenesis is regulated by both VEGF and Ang-Tie signaling pathways. In ACC, early research efforts were focused on VEGF signaling and this pathway was identified as a good prognostic factor and thus a promising therapeutic target. However, every clinical trial so far conducted in ACC using VEGF pathway- targeting drugs, alone or in combination, yielded disappointing results. In contrast, although the Ang-Tie pathway has been pointed out as an important regulator of fetal adrenocortical angiogenesis, its role is yet to be explored in ACC. In the future, further research on the role and efficacy of modulating both Ang-Tie and VEGF pathways in ACC is needed.
Collapse
|
166
|
Guo Z, Wu X, Fan W. Clarifying the effects of diabetes on the cerebral circulation: Implications for stroke recovery and beyond. Brain Res Bull 2021; 171:67-74. [PMID: 33662495 DOI: 10.1016/j.brainresbull.2021.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
Given the sheer increased number of victims per year and the availability of only one effective treatment, acute ischemic stroke (AIS) remains to be one of the most under-treated serious diseases. Diabetes not only increases the incidence of ischemic stroke, but amplifies the ischemic damage, upon which if patients with diabetes suffer from stroke, he/she will confront increased risks of long-term functional deficits. The grim reality makes it a pressing need to intensify efforts at the basic science level to understand how diabetes impairs stroke recovery. This review retrospects the clinical and experimental studies in order to elucidate the detrimental effect of diabetes on cerebrovascular circulation including the major arteries/arterioles, collateral circulation, and neovascularization to shed light on further exploration of novel strategies for cerebral circulation protection before and after AIS in patients with diabetes.
Collapse
Affiliation(s)
- Zhihui Guo
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Xuqing Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Wei Fan
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
167
|
Tie-2, G-CSF, and Leptin as Promising Diagnostic Biomarkers for Endometrial Cancer: A Pilot Study. J Clin Med 2021; 10:jcm10040765. [PMID: 33671851 PMCID: PMC7918088 DOI: 10.3390/jcm10040765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Preoperative determination of the extent of endometrial cancer (EC) would avoid the complications associated with radical surgery. Screening of patients' plasma biomarkers might enable a more precise diagnosis of EC and a tailored treatment approach. This prospective case-control monocentric pilot study included 76 postmenopausal women (38 endometrioid EC patients and 38 control patients with benign gynecological conditions), and 37 angiogenic factors (AFs) were investigated as potential biomarkers for EC. AF concentrations in preoperative plasma samples were measured using Luminex xMAP™ multiplexing technology. The plasma levels of sTie-2 and G-CSF were significantly lower in EC compared to control patients, whereas the plasma levels of leptin were significantly higher in EC patients. Neuropilin-1 plasma levels were significantly higher in patients with type 2 EC (grade 3) compared to patients with lower grade cancer or controls. Follistatin levels were significantly higher in patients with lymphovascular invasion, and IL-8 plasma levels were significantly higher in patients with metastases. If validated, the plasma concentrations of the indicated AFs could represent an important additional diagnostic tool for the early detection and characterization of EC. This could guide the decision-making on the extent of surgery. Further studies with larger patient numbers are currently ongoing.
Collapse
|
168
|
Wu Q, Qi B, Duan X, Ming X, Yan F, He Y, Bu X, Sun S, Zhu H. MicroRNA-126 enhances the biological function of endothelial progenitor cells under oxidative stress via PI3K/Akt/GSK3β and ERK1/2 signaling pathways. Bosn J Basic Med Sci 2021; 21:71-80. [PMID: 31999938 PMCID: PMC7861621 DOI: 10.17305/bjbms.2019.4493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/26/2019] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cell (EPC) transplantation is a safe and effective method to treat acute myocardial infarction (AMI). However, oxidative stress leads to the death of a large number of EPCs in the early stage of transplantation, severely weakening the therapeutic effect. Previous studies demonstrated that microRNAs regulate the biological function of EPCs. The aim of the current study was to investigate the effect of microRNA on the biological function of EPCs under oxidative stress. Quantitative reverse transcription PCR was performed to detect the expression of miR-126, miR-508-5p, miR-150, and miR-16 in EPCs from rats, among which miR-126 showed a relatively higher expression. Treatment with H2O2 decreased miR-126 expression in EPCs in a dose-dependent manner. EPCs were further transfected with miR-126 mimics or inhibitors, followed by H2O2 treatment. Overexpression of miR-126 enhanced the proliferation, migration, and tube formation of H2O2-treated EPCs. MiR-126 overexpression also inhibited reactive oxygen species and malondialdehyde levels and enhanced superoxide dismutase levels, as well as increased angiopoietin (Ang)1 expression and decreased Ang2 expression in H2O2-treated EPCs. Moreover, miR-126 participated in the regulation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in EPCs, where both pathways were activated after miR-126 overexpression in H2O2-treated EPCs. Overall, we showed that miR-126 promoted the biological function of EPCs under H2O2-induced oxidative stress by activating the PI3K/Akt/GSK3β and ERK1/2 signaling pathway, which may serve as a new therapeutic approach to treat AMI.
Collapse
Affiliation(s)
- Qinqin Wu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Benling Qi
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Duan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Ming
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengqin Yan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxia He
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofen Bu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Sun
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
169
|
Tiny miRNAs Play a Big Role in the Treatment of Breast Cancer Metastasis. Cancers (Basel) 2021; 13:cancers13020337. [PMID: 33477629 PMCID: PMC7831489 DOI: 10.3390/cancers13020337] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary MicroRNAs (miRNAs) have emerged as important regulators of tumour progression and metastasis in breast cancer. Through a review of multiple studies, this paper has identified the key regulatory roles of oncogenic miRNAs in breast cancer metastasis including the potentiation of angiogenesis, epithelial-mesenchymal transition, the Warburg effect, and the tumour microenvironment. Several approaches have been studied for selective targeting of breast tumours by miRNAs, ranging from delivery systems such as extracellular vesicles and liposomes to the use of prodrugs and functionally modified vehicle-free miRNAs. While promising, these miRNA-based therapies face challenges including toxicity and immunogenicity, and greater research on their safety profiles must be performed before progressing to clinical trials. Abstract Distant organ metastases accounts for the majority of breast cancer deaths. Given the prevalence of breast cancer in women, it is imperative to understand the underlying mechanisms of its metastatic progression and identify potential targets for therapy. Since their discovery in 1993, microRNAs (miRNAs) have emerged as important regulators of tumour progression and metastasis in various cancers, playing either oncogenic or tumour suppressor roles. In the following review, we discuss the roles of miRNAs that potentiate four key areas of breast cancer metastasis—angiogenesis, epithelial-mesenchymal transition, the Warburg effect and the tumour microenvironment. We then evaluate the recent developments in miRNA-based therapies in breast cancer, which have shown substantial promise in controlling tumour progression and metastasis. Yet, certain challenges must be overcome before these strategies can be implemented in clinical trials.
Collapse
|
170
|
Mathematical simulation of tumour angiogenesis: angiopoietin balance is a key factor in vessel growth and regression. Sci Rep 2021; 11:419. [PMID: 33432093 PMCID: PMC7801613 DOI: 10.1038/s41598-020-79824-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Excessive tumour growth results in a hypoxic environment around cancer cells, thus inducing tumour angiogenesis, which refers to the generation of new blood vessels from pre-existing vessels. This mechanism is biologically and physically complex, with various mathematical simulation models proposing to reproduce its formation. However, although temporary vessel regression is clinically known, few models succeed in reproducing this phenomenon. Here, we developed a three-dimensional simulation model encompassing both angiogenesis and tumour growth, specifically including angiopoietin. Angiopoietin regulates both adhesion and migration between vascular endothelial cells and wall cells, thus inhibiting the cell-to-cell adhesion required for angiogenesis initiation. Simulation results showed a regression, i.e. transient decrease, in the overall length of new vessels during vascular network formation. Using our model, we also evaluated the efficacy of administering the drug bevacizumab. The results highlighted differences in treatment efficacy: (1) earlier administration showed higher efficacy in inhibiting tumour growth, and (2) efficacy depended on the treatment interval even with the administration of the same dose. After thorough validation in the future, these results will contribute to the design of angiogenesis treatment protocols.
Collapse
|
171
|
Campbell KL, Haspel N, Gath C, Kurniatash N, Nouduri Akkiraju I, Stuffers N, Vadher U. Protein hormone fragmentation in intercellular signaling: hormones as nested information systems. Biol Reprod 2021; 104:887-901. [PMID: 33403392 DOI: 10.1093/biolre/ioaa234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 11/14/2022] Open
Abstract
This study explores the hypothesis that protein hormones are nested information systems in which initial products of gene transcription, and their subsequent protein fragments, before and after secretion and initial target cell action, play additional physiological regulatory roles. The study produced four tools and key results: (1) a problem approach that proceeds, with examples and suggestions for in vivo organismal functional tests for peptide-protein interactions, from proteolytic breakdown prediction to models of hormone fragment modulation of protein-protein binding motifs in unrelated proteins; (2) a catalog of 461 known soluble human protein hormones and their predicted fragmentation patterns; (3) an analysis of the predicted proteolytic patterns of the canonical protein hormone transcripts demonstrating near-universal persistence of 9 ± 7 peptides of 8 ± 8 amino acids even after cleavage with 24 proteases from four protease classes; and (4) a coincidence analysis of the predicted proteolysis locations and the 1939 exon junctions within the transcripts that shows an excess (P < 0.001) of predicted proteolysis within 10 residues, especially at the exonal junction (P < 0.01). It appears all protein hormone transcripts generate multiple fragments the size of peptide hormones or protein-protein binding domains that may alter intracellular or extracellular functions by acting as modulators of metabolic enzymes, transduction factors, protein binding proteins, or hormone receptors. High proteolytic frequency at exonal junctions suggests proteolysis has evolved, as a complement to gene exon fusion, to extract structures or functions within single exons or protein segments to simplify the genome by discarding archaic one-exon genes.
Collapse
Affiliation(s)
- Kenneth L Campbell
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Nurit Haspel
- Department of Computer Sciences, University of Massachusetts Boston, Boston, MA, USA
| | - Cassandra Gath
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Nuzulul Kurniatash
- Department of Computer Sciences, University of Massachusetts Boston, Boston, MA, USA
| | | | - Naomi Stuffers
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Uma Vadher
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| |
Collapse
|
172
|
Nosrati H, Aramideh Khouy R, Nosrati A, Khodaei M, Banitalebi-Dehkordi M, Ashrafi-Dehkordi K, Sanami S, Alizadeh Z. Nanocomposite scaffolds for accelerating chronic wound healing by enhancing angiogenesis. J Nanobiotechnology 2021; 19:1. [PMID: 33397416 PMCID: PMC7784275 DOI: 10.1186/s12951-020-00755-7] [Citation(s) in RCA: 290] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/12/2020] [Indexed: 12/23/2022] Open
Abstract
Skin is the body's first barrier against external pathogens that maintains the homeostasis of the body. Any serious damage to the skin could have an impact on human health and quality of life. Tissue engineering aims to improve the quality of damaged tissue regeneration. One of the most effective treatments for skin tissue regeneration is to improve angiogenesis during the healing period. Over the last decade, there has been an impressive growth of new potential applications for nanobiomaterials in tissue engineering. Various approaches have been developed to improve the rate and quality of the healing process using angiogenic nanomaterials. In this review, we focused on molecular mechanisms and key factors in angiogenesis, the role of nanobiomaterials in angiogenesis, and scaffold-based tissue engineering approaches for accelerated wound healing based on improved angiogenesis.
Collapse
Affiliation(s)
- Hamed Nosrati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | | | - Ali Nosrati
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Khodaei
- Department of Materials Science and Engineering, Golpayegan University of Technology, Golpayegan, Iran
| | - Mehdi Banitalebi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Korosh Ashrafi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Sanami
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zohreh Alizadeh
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
173
|
Candelario-Jalil E, Paul S. Impact of aging and comorbidities on ischemic stroke outcomes in preclinical animal models: A translational perspective. Exp Neurol 2021; 335:113494. [PMID: 33035516 PMCID: PMC7874968 DOI: 10.1016/j.expneurol.2020.113494] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/25/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022]
Abstract
Ischemic stroke is a highly complex and devastating neurological disease. The sudden loss of blood flow to a brain region due to an ischemic insult leads to severe damage to that area resulting in the formation of an infarcted tissue, also known as the ischemic core. This is surrounded by the peri-infarct region or penumbra that denotes the functionally impaired but potentially salvageable tissue. Thus, the penumbral tissue is the main target for the development of neuroprotective strategies to minimize the extent of ischemic brain damage by timely therapeutic intervention. Given the limitations of reperfusion therapies with recombinant tissue plasminogen activator or mechanical thrombectomy, there is high enthusiasm to combine reperfusion therapy with neuroprotective strategies to further reduce the progression of ischemic brain injury. Till date, a large number of candidate neuroprotective drugs have been identified as potential therapies based on highly promising results from studies in rodent ischemic stroke models. However, none of these interventions have shown therapeutic benefits in stroke patients in clinical trials. In this review article, we discussed the urgent need to utilize preclinical models of ischemic stroke that more accurately mimic the clinical conditions in stroke patients by incorporating aged animals and animal stroke models with comorbidities. We also outlined the recent findings that highlight the significant differences in stroke outcome between young and aged animals, and how major comorbid conditions such as hypertension, diabetes, obesity and hyperlipidemia dramatically increase the vulnerability of the brain to ischemic damage that eventually results in worse functional outcomes. It is evident from these earlier studies that including animal models of aging and comorbidities during the early stages of drug development could facilitate the identification of neuroprotective strategies with high likelihood of success in stroke clinical trials.
Collapse
Affiliation(s)
- Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
174
|
Pereira M, Matuszewska K, Jamieson C, Petrik J. Characterizing Endocrine Status, Tumor Hypoxia and Immunogenicity for Therapy Success in Epithelial Ovarian Cancer. Front Endocrinol (Lausanne) 2021; 12:772349. [PMID: 34867818 PMCID: PMC8635771 DOI: 10.3389/fendo.2021.772349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer is predominantly diagnosed at advanced stages which creates significant therapeutic challenges. As a result, the 5-year survival rate is low. Within ovarian cancer, significant tumor heterogeneity exists, and the tumor microenvironment is diverse. Tumor heterogeneity leads to diversity in therapy response within the tumor, which can lead to resistance or recurrence. Advancements in therapy development and tumor profiling have initiated a shift from a "one-size-fits-all" approach towards precision patient-based therapies. Here, we review aspects of ovarian tumor heterogeneity that facilitate tumorigenesis and contribute to treatment failure. These tumor characteristics should be considered when designing novel therapies or characterizing mechanisms of treatment resistance. Individual patients vary considerably in terms of age, fertility and contraceptive use which innately affects the endocrine milieu in the ovary. Similarly, individual tumors differ significantly in their immune profile, which can impact the efficacy of immunotherapies. Tumor size, presence of malignant ascites and vascular density further alters the tumor microenvironment, creating areas of significant hypoxia that is notorious for increasing tumorigenesis, resistance to standard of care therapies and promoting stemness and metastases. We further expand on strategies aimed at improving oxygenation status in tumors to dampen downstream effects of hypoxia and set the stage for better response to therapy.
Collapse
|
175
|
Jin Y, Ji W, Yang H, Chen S, Zhang W, Duan G. Endothelial activation and dysfunction in COVID-19: from basic mechanisms to potential therapeutic approaches. Signal Transduct Target Ther 2020; 5:293. [PMID: 33361764 PMCID: PMC7758411 DOI: 10.1038/s41392-020-00454-7] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
On 12 March 2020, the outbreak of coronavirus disease 2019 (COVID-19) was declared a pandemic by the World Health Organization. As of 4 August 2020, more than 18 million confirmed infections had been reported globally. Most patients have mild symptoms, but some patients develop respiratory failure which is the leading cause of death among COVID-19 patients. Endothelial cells with high levels of angiotensin-converting enzyme 2 expression are major participants and regulators of inflammatory reactions and coagulation. Accumulating evidence suggests that endothelial activation and dysfunction participate in COVID-19 pathogenesis by altering the integrity of vessel barrier, promoting pro-coagulative state, inducing endothelial inflammation, and even mediating leukocyte infiltration. This review describes the proposed cellular and molecular mechanisms of endothelial activation and dysfunction during COVID-19 emphasizing the principal mediators and therapeutic implications.
Collapse
Affiliation(s)
- Yuefei Jin
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Wangquan Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Haiyan Yang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Shuaiyin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Weiguo Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
| |
Collapse
|
176
|
Bijkerk R, Kallenberg MH, Zijlstra LE, van den Berg BM, de Bresser J, Hammer S, Bron EE, Achterberg H, van Buchem MA, Berkhout-Byrne NC, Bos WJW, van Heemst D, Rabelink TJ, van Zonneveld AJ, van Buren M, Mooijaart S. Circulating angiopoietin-2 and angiogenic microRNAs associate with cerebral small vessel disease and cognitive decline in older patients reaching end stage renal disease. Nephrol Dial Transplant 2020; 37:498-506. [PMID: 33355649 DOI: 10.1093/ndt/gfaa370] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The prevalence of end-stage renal disease (ESRD) is increasing worldwide, with the majority of new ESRD cases diagnosed in patients aged >60 years. These older patients are at increased risk for impaired cognitive functioning, potentially through cerebral small vessel disease (SVD). Novel markers of vascular integrity may be of clinical value for identifying patients at high risk for cognitive impairment. METHODS We aimed to associate the levels of Angiopoietin-2 (Ang-2), asymmetric dimethylarginine (ADMA), and a selection of eight circulating angiogenic miRNAs with SVD and cognitive impairment in older patients reaching ESRD that did not initiate renal replacement therapy yet (n = 129; mean age 75.3 years; mean eGFR 16.4 mL/min). We assessed brain MRI changes of SVD (white matter hyperintensity volume, microbleeds and presence of lacunes) and measures of cognition in domains of memory, psychomotor speed and executive function, comprised in a neuropsychological test battery. RESULTS Older patients reaching ESRD showed an unfavorable angiogenic profile, as indicated by aberrant levels of Ang-2 and five angiogenic miRNAs (miR-27a, miR-126, miR-132, miR-223, miR-326), compared to healthy persons and patients with diabetic nephropathy. Moreover, Ang-2 associated with SVD and with the domains of psychomotor speed and executive function, while miR-223 and miR-29a associated with memory function. CONCLUSIONS Taken together, these novel angiogenic markers might serve to identify older patients with ESRD at risk of cognitive decline, as well as give insight into the underlying (vascular) pathophysiology.
Collapse
Affiliation(s)
- Roel Bijkerk
- Department of Internal Medicine (Nephrology).,Einthoven Laboratory for Vascular and Regenerative Medicine
| | - Marije H Kallenberg
- Department of Internal Medicine (Nephrology).,Department of Internal Medicine (Gerontology and Geriatrics)
| | - Laurien E Zijlstra
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine (Nephrology).,Einthoven Laboratory for Vascular and Regenerative Medicine
| | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Esther E Bron
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Hakim Achterberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Willem Jan W Bos
- Department of Internal Medicine (Nephrology).,Department of Internal Medicine, St Antonius Hospital, Nieuwegein, The Netherlands
| | | | - Ton J Rabelink
- Department of Internal Medicine (Nephrology).,Einthoven Laboratory for Vascular and Regenerative Medicine
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology).,Einthoven Laboratory for Vascular and Regenerative Medicine
| | - Marjolijn van Buren
- Department of Internal Medicine (Nephrology).,Department of Nephrology, HAGA Hospital, The Hague, The Netherlands
| | - Simon Mooijaart
- Department of Internal Medicine (Gerontology and Geriatrics).,Institute of Evidence-Based Medicine in Old Age, Leiden, The Netherlands
| |
Collapse
|
177
|
Proanthocyanidins Should Be a Candidate in the Treatment of Cancer, Cardiovascular Diseases and Lipid Metabolic Disorder. Molecules 2020; 25:molecules25245971. [PMID: 33339407 PMCID: PMC7766935 DOI: 10.3390/molecules25245971] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
The conventional view of using medicines as routine treatment of an intractable disease is being challenged in the face of extensive and growing evidence that flavonoids in foods, especially proanthocyanidins (PAs), can participate in tackling fatal diseases like cancer, cardiovascular and lipid metabolic diseases, both as a precautionary measure or as a dietary treatment. Although medical treatment with medicines will remain necessary in some cases, at least in the short term, PAs’ function as antioxidant, anti-inflammatory drugs, signal pathway regulators remain critical in many diseases. This review article demonstrates the physical and biological properties of PAs, summarizes the health benefits of PAs found by researchers previously, and shows the possibility and importance of being a dietary treatment substance.
Collapse
|
178
|
Determining the Value of Angiopoietin-2 Biomarker in Predicting Complications of Leptospirosis. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2020. [DOI: 10.5812/archcid.87162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Leptospirosis is a common zoonosis disease that is potentially life-threatening all around the world. Angiopoietin-2 can be used as an indicator to determine the severity of the disease in critically ill patients. Objectives: This study aimed to determine the value of angiopoietin-2 biomarker in predicting leptospirosis complications. Methods: In this prospective observational study, 90 participants, including 45 patients with leptospirosis and 45 participants in the control group, are studied. Three to five days after the onset of symptoms before starting treatment with antibiotics, 5 mL venous blood was taken to measuring the angiopoietin-2 level by enzyme-linked immunosorbent assay (ELISA). Results: In this study, 75 men (83.3%) and 15 women (16.7%) with an age distribution of 23 to 75 years participated. A total of 74 (82.3%) patients were employed in the agriculture sector, while 59 (65.6%) lived in rural areas. There was a significant relationship between patients with severe forms of leptospirosis and levels of angiopoietin-2 (P-value < 0.01). Also, there was a significant relationship between angiopoietin-2 biomarker level and markers of disease severity such as anemia (P-value < 0.01), oliguria (P-value < 0.01), severe thrombocytopenia (P-value < 0.01), length of stay (P-value < 0.01), and CRP more than 2+ (P-value < 0.001). Conclusions: This study demonstrated that angiopoietin-2 biomarker can be used in screening disease severity and predicting complications of leptospirosis.
Collapse
|
179
|
Di Matteo A, Belloni E, Pradella D, Cappelletto A, Volf N, Zacchigna S, Ghigna C. Alternative splicing in endothelial cells: novel therapeutic opportunities in cancer angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:275. [PMID: 33287867 PMCID: PMC7720527 DOI: 10.1186/s13046-020-01753-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Alternative splicing (AS) is a pervasive molecular process generating multiple protein isoforms, from a single gene. It plays fundamental roles during development, differentiation and maintenance of tissue homeostasis, while aberrant AS is considered a hallmark of multiple diseases, including cancer. Cancer-restricted AS isoforms represent either predictive biomarkers for diagnosis/prognosis or targets for anti-cancer therapies. Here, we discuss the contribution of AS regulation in cancer angiogenesis, a complex process supporting disease development and progression. We consider AS programs acting in a specific and non-redundant manner to influence morphological and functional changes involved in cancer angiogenesis. In particular, we describe relevant AS variants or splicing regulators controlling either secreted or membrane-bound angiogenic factors, which may represent attractive targets for therapeutic interventions in human cancer.
Collapse
Affiliation(s)
- Anna Di Matteo
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Elisa Belloni
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Davide Pradella
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Ambra Cappelletto
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Nina Volf
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy. .,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149, Trieste, Italy.
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
180
|
Isaji T, Osuka K, Ohmichi Y, Ohmichi M, Naito M, Nakano T, Iwami K, Miyachi S. Expression of Angiopoietins and Angiogenic Signaling Pathway Molecules in Chronic Subdural Hematomas. J Neurotrauma 2020; 37:2493-2498. [DOI: 10.1089/neu.2020.7042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Taiki Isaji
- Department of Neurosurgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Koji Osuka
- Department of Neurosurgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yusuke Ohmichi
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Mika Ohmichi
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Munekazu Naito
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takashi Nakano
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kenichiro Iwami
- Department of Neurosurgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Shigeru Miyachi
- Department of Neurosurgery, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
181
|
Forma A, Tyczyńska M, Kędzierawski P, Gietka K, Sitarz M. Gastric carcinogenesis: a comprehensive review of the angiogenic pathways. Clin J Gastroenterol 2020; 14:14-25. [PMID: 33206367 PMCID: PMC7886717 DOI: 10.1007/s12328-020-01295-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is undoubtedly one of the most prevalent malignancies worldwide. Since GC is the second leading cause of cancer-related deaths with nearly one million new diagnoses reported every year, there is a need for the development of new, effective treatment strategies of GC. Gastric carcinogenesis is a complex process that is induced by numerous factors and further stimulated by many pro-oncogenic pathways. Angiogenesis is the process of the new blood vessels formation from the already existing ones and it significantly contributes to the progression of gastric tumorigenesis and the growth of the cancerous tissues. The newly formed vessels provide cancer cells with proper nutrition, growth factors, and oxygen supply that are crucial for tumor growth and progression. Tumor-associated vessels differ from the physiological ones both morphologically and functionally. They are usually inefficient and unevenly distributed due to structural transformations. Thus, the development of the angiogenesis inhibitors that possess therapeutic effects has been the main focus of recent studies. Angiogenesis inhibitors mostly affect the vascular endothelial growth factor (VEGF) pathway since it is a major factor that stimulates the pro-angiogenic pathways. The aim of this review was to describe and summarize other promising molecular pathways that might be crucial in further improvements in GC therapies. This article provides an overview of how a meaningful role in tumor progression the angiogenetic process has. Furthermore, this review includes a description of the most important angiogenic factors as well as pathways and their involvement in gastric carcinogenesis.
Collapse
Affiliation(s)
- Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland.
| | - Magdalena Tyczyńska
- Department of Human Anatomy, Medical University of Lublin, 20-090, Lublin, Poland
| | - Paweł Kędzierawski
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Klaudyna Gietka
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090, Lublin, Poland
| |
Collapse
|
182
|
Plazak ME, Hankinson SJ, Sorensen EN, Reed BN, Ravichandran B, Ton VK. Novel Use of Tamoxifen to Reduce Recurrent Gastrointestinal Bleeding in Patients with Left Ventricular Assist Devices. J Cardiovasc Transl Res 2020; 14:484-491. [PMID: 33175315 DOI: 10.1007/s12265-020-10084-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/03/2020] [Indexed: 11/30/2022]
Abstract
Gastrointestinal bleeding (GIB) is a frequent complication in patients with continuous-flow left ventricular assist devices (LVAD). We retrospectively evaluated eight patients implanted with a HeartWare LVAD between July 2017 and June 2020 who experienced at least one episode of GIB and were started on tamoxifen 20 mg once daily for secondary prevention. Tamoxifen was associated with a significant decrease in major GIB from a median of 3 (IQR 1.4-7) events/patient-year pre-tamoxifen initiation to 0 (IQR 0-0.9) events/patient-year post-tamoxifen initiation (p = 0.02). Transfusion of packed red blood cells also decreased from 16.8 (IQR 9.9-30.6) units/patient-year pre-tamoxifen initiation to 1.5 (IQR 0-7.5) units/patient-year post-tamoxifen (p = 0.04). Tamoxifen was well tolerated and no thromboembolic complications were observed. This small cohort study suggests that tamoxifen is associated with reduced GIB and transfusion requirements, with no apparent increase in thrombotic risk. A larger, randomized study is warranted to confirm the results of this exploratory analysis. Graphical abstract.
Collapse
Affiliation(s)
- Michael E Plazak
- University of Maryland Medical Center, 22 South Greene Street, Suite 400, Baltimore, MD, 21201, USA.
| | - Stephen J Hankinson
- Department of Medicine, Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik N Sorensen
- Division of Perioperative Services, University of Maryland Medical Center, Baltimore, MD, USA
| | - Brent N Reed
- Department of Pharmacy Practice and Science, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Bharath Ravichandran
- University of Maryland Medical Center, 22 South Greene Street, Suite 400, Baltimore, MD, 21201, USA
| | - Van-Khue Ton
- Department of Medicine, Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Harvard Medical School, Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
183
|
Cataldo R, De Nunzio G, Millithaler JF, Alfinito E. Aptamers Which Target Proteins: What Proteotronics Suggests to Pharmaceutics. Curr Pharm Des 2020; 26:363-371. [PMID: 31942851 DOI: 10.2174/1381612826666200114095027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/08/2020] [Indexed: 01/01/2023]
Abstract
Aptamers represent a challenging field of research, relevant for diagnosis in macular degeneration, cancer, thrombosis and many inflammatory diseases, and promising in drug discovery and development. Their selection is currently performed by a stable in vitro technology, namely, SELEX. Furthermore, computationalstatistical tools have been developed to complement the SELEX selection; they work both in the preliminary stage of selection, by designing high affinity aptamers for the assigned target, and also in the final stage, analyzing the features of the best performers to implement the selection technique further. A massive use of the in silico approach is, at present, only restricted by the limited knowledge of the specific aptamer-target topology. Actually, only about fifty X-ray structures of aptamer-protein complexes have been experimentally resolved, highlighting how this knowledge has to be improved. The structure of biomolecules like aptamer-protein complexes can be represented by networks, from which several parameters can be extracted. This work briefly reviews the literature, discussing if and how general network parameters in the framework of Proteotronics and graph theory (such as electrical features, link number, free energy change, and assortativity), are important in characterizing the complexes, anticipating some features of the biomolecules. To better explain this topic, a case-study is proposed, constituted by a set of anti-angiopoietin (Ang2) aptamers, whose performances are known from the experiments, and for which two different types of conformers were predicted. A topological indicator is proposed, named Möbius (M), which combines local and global information, and seems able to discriminate between the two possible types of conformers, so that it can be considered as a useful complement to the in vitro screening for pharmaceutical aims.
Collapse
Affiliation(s)
- Rosella Cataldo
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, Lecce, Italy.,Laboratory of Interdisciplinary Research Applied to Medicine (DReAM), University of Salento and ASL (Local Health Authority), Lecce, Italy
| | - Giorgio De Nunzio
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, Lecce, Italy.,Laboratory of Interdisciplinary Research Applied to Medicine (DReAM), University of Salento and ASL (Local Health Authority), Lecce, Italy
| | - Jean-Francois Millithaler
- Electrical and Computer Engineering, University of Massachusetts Lowell, Lowell, MA, 01854, United States
| | - Eleonora Alfinito
- Department of Innovation Engineering, University of Salento, Lecce, Italy
| |
Collapse
|
184
|
Zhong J, Lu W, Zhang J, Huang M, Lyu W, Ye G, Deng L, Chen M, Yao N, Li Y, Liu G, Liang Y, Fu J, Zhang D, Ye W. Notoginsenoside R1 activates the Ang2/Tie2 pathway to promote angiogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153302. [PMID: 32823242 DOI: 10.1016/j.phymed.2020.153302] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/15/2020] [Accepted: 08/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Therapeutic angiogenesis is a novel strategy for the treatment of ischemic diseases that involves promotion of angiogenesis in ischemic tissues via the use of proangiogenic agents. However, effective proangiogenic drugs that activate the Ang2/Tie2 signaling pathway remain scarce. PURPOSE We aimed to investigate the proangiogenic activity of notoginsenoside R1 (NR1) isolated from total saponins of Panax notoginseng with regard to activation of the Ang2/Tie2 signaling pathway. METHODS We examined the proangiogenic effects of NR1 by assessing the effects of NR1 on the proliferation, migration, invasion and tube formation of human umbilical vein endothelial cells (HUVECs). The aortic ring assay and vascular endothelial growth factor receptor inhibitor (VRI)-induced vascular regression in the zebrafish model were used to confirm the proangiogenic effects of NR1 ex vivo and in vivo. Furthermore, the molecular mechanism was investigated by Western blot analysis. RESULTS We found that NR1 promoted the proliferation, mobility and tube formation of HUVECs in vitro. NR1 also increased the number of sprouting vessels in rat aortic rings and rescued VRI-induced vascular regression in zebrafish. NR1-induced angiogenesis was dependent on Tie2 receptor activation mediated by increased autocrine Ang2 in HUVECs, and inhibition of the Ang2/Tie2 pathway abrogated the proangiogenic effects of NR1. CONCLUSIONS Our results suggest that NR1 promotes angiogenesis by activating the Ang2/Tie2 signaling pathway. Thus, NR1-induced activation of the Ang2/Tie2 pathway is an effective proangiogenic approach. NR1 may be useful agent for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Jincheng Zhong
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Weijin Lu
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Jiayan Zhang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Wenyu Lyu
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Geni Ye
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Lijuan Deng
- Formula‑pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Nan Yao
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Guanping Liu
- Guangxi Engineering Research Center of Innovative Preparations for Natural Medicine, Guangxi Wuzhou Pharmaceutical (Group) Co., Ltd, Wuzhou 543000, China
| | - Yunfei Liang
- Guangxi Engineering Research Center of Innovative Preparations for Natural Medicine, Guangxi Wuzhou Pharmaceutical (Group) Co., Ltd, Wuzhou 543000, China
| | - Jingwen Fu
- The Affiliated High School of South China Normal University, Guangzhou 510630, China
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China.
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
185
|
Lee EJ, Jain M, Alimperti S. Bone Microvasculature: Stimulus for Tissue Function and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:313-329. [PMID: 32940150 DOI: 10.1089/ten.teb.2020.0154] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone is a highly vascularized organ, providing structural support to the body, and its development, regeneration, and remodeling depend on the microvascular homeostasis. Loss or impairment of vascular function can develop diseases, such as large bone defects, avascular necrosis, osteoporosis, osteoarthritis, and osteopetrosis. In this review, we summarize how vasculature controls bone development and homeostasis in normal and disease cases. A better understanding of this process will facilitate the development of novel disease treatments that promote bone regeneration and remodeling. Specifically, approaches based on tissue engineering components, such as stem cells and growth factors, have demonstrated the capacity to induce bone microvasculature regeneration and mineralization. This knowledge will have relevant clinical implications for the treatment of bone disorders by developing novel pharmaceutical approaches and bone grafts. Finally, the tissue engineering approaches incorporating vascular components may widely be applied to treat other organ diseases by enhancing their regeneration capacity. Impact statement Bone vasculature is imperative in the process of bone development, regeneration, and remodeling. Alterations or disruption of the bone vasculature leads to loss of bone homeostasis and the development of bone diseases. In this study, we review the role of vasculature on bone diseases and how vascular tissue engineering strategies, with a detailed emphasis on the role of stem cells and growth factors, will contribute to bone therapeutics.
Collapse
Affiliation(s)
- Eun-Jin Lee
- American Dental Association Science and Research Institute, Gaithersburg, Maryland, USA
| | - Mahim Jain
- Kennedy Krieger Institute, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stella Alimperti
- American Dental Association Science and Research Institute, Gaithersburg, Maryland, USA
| |
Collapse
|
186
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
187
|
Koul A, Bansal MP, Aniqa A, Chaudhary H, Chugh NA. Lycopene enriched tomato extract suppresses chemically induced skin tumorigenesis in mice. INT J VITAM NUTR RES 2020; 90:493-513. [DOI: 10.1024/0300-9831/a000597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract. The present study revealed the effects of Lycopene enriched tomato extract (LycT) on chemically induced skin cancer in mice. Skin tumors were induced by topical application of 7,12-Dimethylbenz(a)anthracene (DMBA) [500 nmol/100 ul of acetone, twice a week for two weeks] and 12-O-tetradecanoyl phorbol-13-acetate (TPA) [1.7 nmol/100 ul of acetone, twice a week for eighteen weeks] and LycT (5 mg/kg b.w.) was administered orally. Male Balb/c mice were divided into four groups (n = 15 per group): control, DMBA/TPA, LycT and LycT + DMBA/TPA. The chemopreventive response of LycT to skin tumorigenesis was evident by inhibition in tumor incidence, number, size, burden and volume in LycT + DMBA/TPA group when compared to DMBA/TPA group. This was associated with inhibition of cell proliferation in LycT + DMBA/TPA group as observed by the decrease in epidermal morphometric parameters and mRNA and protein expression of proliferating cell nuclear antigen when compared to DMBA/TPA group (p ≤ 0.05). LycT decreased (p ≤ 0.05) the mRNA and protein expression of angiogenic genes (vascular endothelial growth factor, angiopoietin-2, basic fibroblast growth factor) in LycT + DMBA/TPA group, suggesting its anti-angiogenic effects. The increase (p ≤ 0.05) in protein expression of connexin-32 and 43 in LycT + DMBA/TPA group suggests improved inter cellular communication when compared to DMBA/TPA group. Histochemical studies demonstrated that the components of extracellular matrix (fibrous proteins and mucopolysaccharides) were also modulated during skin carcinogenesis and its chemoprevention by LycT. The decrease in cell proliferation parameters and expression of angiogenesis associated genes, modulation of ECM components and increase in expression of connexins suggest that LycT improved multiple dysregulated processes during chemoprevention of skin cancer.
Collapse
Affiliation(s)
- Ashwani Koul
- Department of Biophysics, Panjab University, Chandigarh, India
| | | | - Aniqa Aniqa
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Harsh Chaudhary
- Department of Biophysics, Panjab University, Chandigarh, India
| | | |
Collapse
|
188
|
Martin-Liberal J, Hollebecque A, Aftimos P, Jungels C, Martin-Romano P, Rodon J, Kremer JD, Zhang W, Bendell J. First-in-human, dose-escalation, phase 1 study of anti-angiopoietin-2 LY3127804 as monotherapy and in combination with ramucirumab in patients with advanced solid tumours. Br J Cancer 2020; 123:1235-1243. [PMID: 32741971 PMCID: PMC7555513 DOI: 10.1038/s41416-020-1011-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND This is the first-in-human study of novel anti-angiopoietin-2 (Ang-2) monoclonal antibody LY3127804 as monotherapy and in combination with ramucirumab in advanced solid tumours. METHODS Patients received intravenous LY3127804 monotherapy (4, 8, 12, 16, 20 and 27 mg/kg) in part A; LY3127804 (8, 12, 16, 20 and 27 mg/kg) with 8 mg/kg ramucirumab in part B; and LY3127804 (20 mg/kg) with 12 mg/kg ramucirumab in part C. Treatments were administered every 2 weeks (Q2W) during 28-day cycles. Dose-escalation was based on cycle 1 dose-limiting toxicities (DLTs). RESULTS Sixty-two patients were treated in part A (n = 20), part B (n = 35) and part C (n = 7). Constipation, diarrhoea and fatigue were the most common treatment-emergent adverse events (TEAEs) in part A; hypertension and peripheral oedema were the most frequent TEAE in parts B and C. No DLT was observed and maximum tolerated dose for LY3127804 was not reached. Four patients achieved partial response with combination therapy (clear cell endometrial carcinoma, cervix squamous cell carcinoma, carcinoma of unknown primary and gastroesophageal junction carcinoma), 29 achieved stable disease, and 24 had progressive disease. CONCLUSIONS LY3127804 monotherapy and its combination with ramucirumab are well tolerated. LY3127804 20 mg/kg was the recommended Phase 2 dose.
Collapse
Affiliation(s)
- Juan Martin-Liberal
- Department of Medical Oncology, Vall d'Hebrón Institute of Oncology (VHIO), Barcelona, Spain.
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Barcelona, Spain.
| | | | - Philippe Aftimos
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christiane Jungels
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Jordi Rodon
- Department of Medical Oncology, Vall d'Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Wei Zhang
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Johanna Bendell
- Department of Medical Oncology, Sarah Cannon Research Institute, Nashville, TN, USA
| |
Collapse
|
189
|
Lu X. Structure and Function of Angiopoietin-like Protein 3 (ANGPTL3) in Atherosclerosis. Curr Med Chem 2020; 27:5159-5174. [PMID: 31223079 DOI: 10.2174/0929867326666190621120523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Angiopoietin-Like Proteins (ANGPTLs) are structurally related to the angiopoietins. A total of eight ANGPTLs (from ANGPTL1 to ANGPTL8) have been identified so far. Most ANGPTLs possess multibiological functions on lipid metabolism, atherosclerosis, and cancer. Among them, ANGPTL3 has been shown to regulate the levels of Very Low-Density Lipoprotein (VLDL) made by the liver and play a crucial role in human lipoprotein metabolism. METHOD A systematic appraisal of ANGPTLs was conducted, focusing on the main features of ANGPTL3 that has a significant role in atherosclerosis. RESULTS Angiopoietins including ANGPTL3 are vascular growth factors that are highly specific for endothelial cells, perform a variety of other regulatory activities to influence inflammation, and have been shown to possess both pro-atherosclerotic and atheroprotective effects. CONCLUSION ANGPTL3 has been demonstrated as a promising target in the pharmacological management of atherosclerosis. However, many questions remain about its biological functions.
Collapse
Affiliation(s)
- Xinjie Lu
- The Mary and Garry Weston Molecular Immunology Laboratory, Thrombosis Research Institute, London SW3 6LR, England, United Kingdom
| |
Collapse
|
190
|
Valentini G, Pope JE. Undifferentiated connective tissue disease at risk for systemic sclerosis: Which patients might be labeled prescleroderma? Autoimmun Rev 2020; 19:102659. [PMID: 32942034 DOI: 10.1016/j.autrev.2020.102659] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/12/2020] [Indexed: 02/06/2023]
Abstract
Undifferentiated Connective Tissue Disease at risk for Systemic Sclerosis (UCTD-risk-SSc), otherwise referred to as very early-early SSc (very early-early diagnosis of systemic sclerosis VEDOSS), is a condition characterized by Raynaud's phenomenon (RP) and either SSc serum marker autoantibodies or a capillaroscopic scleroderma pattern or both, but without satisfying classification criteria for SSc neither features consistent with SSc sine scleroderma. Approximately half the UCTD-risk-SSc patients develop definite SSc over 5-10 years of follow-up. Identifying patients who will undergo such evolution is an unmet need. Predicting at onset which patients with RP are going to develop SSc over time has long been a research objective and still is an unaccomplished task. The present review is devoted to the critical analysis of the nosographic boundaries of the condition and of items predictive of evolution including serological, capillaroscopic and circulating markers. A weighted score, based on serum antinuclear antibody titre, serum marker antibodies positivity and avascular areas has been developed and may identify in the meanwhile patients to be labeled prescleroderma i.e. those probably developing SSc over time. Future research should be directed to investigate unexplored features, validate and improve the performance of the score and highlight the involved pathways to be contrasted in order to identify a targeted therapy hampering the development of overt SSc.
Collapse
Affiliation(s)
- Gabriele Valentini
- Università degli Studi della Campania "Luigi Vanvitelli", Department of Precision Medicine, Section of Rheumatology, Italy.
| | - Janet E Pope
- Università degli Studi della Campania "Luigi Vanvitelli", Department of Precision Medicine, Section of Rheumatology, Italy; Schulich School of Medicine and Dentistry, University of Western Ontario, Division of Rheumatology, St. Joseph's Health Care, London, Ontario, Canada
| |
Collapse
|
191
|
Valipour E, Nooshabadi VT, Mahdipour S, Shabani S, Farhady-Tooli L, Majidian S, Noroozi Z, Mansouri K, Motevaseli E, Modarressi MH. Anti-angiogenic effects of testis-specific gene antigen 10 on primary endothelial cells. Gene 2020; 754:144856. [PMID: 32512160 DOI: 10.1016/j.gene.2020.144856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/24/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023]
Abstract
Growing evidence indicates the antitumor and antiangiogenesis activities of testis-specific gene antigen 10 (TSGA10). However, the underlying mechanisms and precise role of TSGA10 in angiogenesis are still elusive. In this study, we isolated human umbilical cord vein endothelial cells (HUVECs) and stably transfected with pcDNA3.1 carrying TSGA10 coding sequence. We demonstrated that TSGA10 over-expression significantly decreases HUVEC tubulogenesis and interconnected capillary network formation. HUVECs over-expressing TSGA10 exhibited a significant decrease in migration and proliferation rates. TSGA10 over-expression markedly decreased expression of angiogenesis-related genes, including VEGF-A, VEGFR-2, Ang-1, Ang-2, and Tie-2. Our ELISA results showed the decrease in VEGF-A mRNA expression level is associated with a significant decrease in its protein secretion. Additionally, over-expressing TSGA10 decreased expression levels of marker genes of cell migration (MMP-2, MMP-9, and SDF-1a) and proliferation (PCNA and Ki-67. Furthermore, ERK-1 and AKT phosphorylation significantly reduced in HUVECs over-expressing TSGA10. Our findings suggest a potent anti-angiogenesis activity of TSGA10 in HUVECs through down-regulation of ERK and AKT signalling pathways, and may provide therapeutic benefits for the management of different pathological angiogenesis.
Collapse
Affiliation(s)
- Elahe Valipour
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Shadi Mahdipour
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sasan Shabani
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Farhady-Tooli
- Department of Microbiology, School of Biology, College of Science, Tehran University, Tehran, Iran
| | - Sina Majidian
- School of Electrical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Zahra Noroozi
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elaheh Motevaseli
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
192
|
Acıkgoz Mert GS, Ceri M, Calli Demirkan N, Sahin B, Mert M, Dursun B. Effect of bevacizumab and everolimus combination treatment on peritoneal sclerosis in an experimental rat model. Ther Apher Dial 2020; 25:323-330. [PMID: 33439548 DOI: 10.1111/1744-9987.13565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 10/23/2022]
Abstract
The aim of this study was to investigate whether bevacizumab and everolimus combination therapy is superior to bevacizumab treatment alone as a treatment for peritoneal sclerosis. Forty Wistar albino rats were divided into five equal groups. The control group received isotonic saline solution (2 mL/day) intraperitoneal (IP) daily for 3 weeks. The CG group received 2 mL 0.1% chlorhexidine gluconate (CG) and 15% ethanol dissolved in saline IP daily for 3 weeks. Peritoneal tissue samples were taken at the end of 3 weeks. The resting group received CG (weeks 0-3), plus isotonic saline solution (2 mL/day) IP daily and tap water (2 mL/day) via a feeding tube daily (weeks 3-6).The bevacizumab group received CG (weeks 1-3) plus bevacizumab at 2.5 mg/kg/day (2 mL) IP daily and tap water (2 mL/day) via a feeding tube daily (weeks 3-6). The bevacizumab+everolimus group received CG (weeks 1-3) plus bevacizumab at 2.5 mg/kg/day (2 mL) IP daily and everolimus at 0.3 mg/kg/day (2 mL) via a feeding tube daily (weeks 3-6). Peritoneal tissue samples were taken from these three groups at the end of 6 weeks and were examined after staining with hematoxylin-eosin and Masson's trichrome. Inflammation, vasculopathy, fibrosis, and peritoneal thickness were evaluated under light microscopy. The samples were also stained with anti-TGF-β and anti-MMP-2. Inflammation and vasculopathy scores were significantly decreased in the VEGF-i group compared to the CG group. The addition of everolimus to VEGF-i showed significantly lower inflammation, vasculopathy, fibrosis scores, and an evident decrease in peritoneal thickening (respectively, 2.29 ± 0.76 vs 0.57 ± 0.53, P = .003; 2.71 ± 0.76 vs 1.43 ± 0.53, P = .008; 2.57 ± 0.79 vs 1.57 ± 0.79, P = .04; 247.5 ± 136.1 vs 84.5 ± 48.6, P = .048). MMP-2 levels were lower in the combination group compared to the resting group (2.63 ± 0.74 vs 1.86 ± 0.38, P = .019). The study results demonstrated that bevacizumab and everolimus combination therapy was more effective than bevacizumab therapy alone.
Collapse
Affiliation(s)
| | - Mevlut Ceri
- Department of Nephrology, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Nese Calli Demirkan
- Department of Pathology, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Barbaros Sahin
- Department of Experimental Surgery Application and Research, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Mehmet Mert
- Department of Internal Medicine, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Belda Dursun
- Department of Nephrology, Pamukkale University Faculty of Medicine, Denizli, Turkey
| |
Collapse
|
193
|
Wu SZ, Roden DL, Wang C, Holliday H, Harvey K, Cazet AS, Murphy KJ, Pereira B, Al-Eryani G, Bartonicek N, Hou R, Torpy JR, Junankar S, Chan CL, Lam CE, Hui MN, Gluch L, Beith J, Parker A, Robbins E, Segara D, Mak C, Cooper C, Warrier S, Forrest A, Powell J, O'Toole S, Cox TR, Timpson P, Lim E, Liu XS, Swarbrick A. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J 2020; 39:e104063. [PMID: 32790115 DOI: 10.15252/embj.2019104063] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/30/2022] Open
Abstract
The tumour stroma regulates nearly all stages of carcinogenesis. Stromal heterogeneity in human triple-negative breast cancers (TNBCs) remains poorly understood, limiting the development of stromal-targeted therapies. Single-cell RNA sequencing of five TNBCs revealed two cancer-associated fibroblast (CAF) and two perivascular-like (PVL) subpopulations. CAFs clustered into two states: the first with features of myofibroblasts and the second characterised by high expression of growth factors and immunomodulatory molecules. PVL cells clustered into two states consistent with a differentiated and immature phenotype. We showed that these stromal states have distinct morphologies, spatial relationships and functional properties in regulating the extracellular matrix. Using cell signalling predictions, we provide evidence that stromal-immune crosstalk acts via a diverse array of immunoregulatory molecules. Importantly, the investigation of gene signatures from inflammatory-CAFs and differentiated-PVL cells in independent TNBC patient cohorts revealed strong associations with cytotoxic T-cell dysfunction and exclusion, respectively. Such insights present promising candidates to further investigate for new therapeutic strategies in the treatment of TNBCs.
Collapse
Affiliation(s)
- Sunny Z Wu
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel L Roden
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Chenfei Wang
- Department of Data Sciences, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Holly Holliday
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Kate Harvey
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Aurélie S Cazet
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Kendelle J Murphy
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Brooke Pereira
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Ghamdan Al-Eryani
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Nenad Bartonicek
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA, Australia
| | - James R Torpy
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Simon Junankar
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Chia-Ling Chan
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Chuan En Lam
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Mun N Hui
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Laurence Gluch
- The Strathfield Breast Centre, Strathfield, NSW, Australia
| | - Jane Beith
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | | | | | | | - Cindy Mak
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Caroline Cooper
- Pathology Queensland, Princess Alexandra Hospital, Brisbane, Qld, Australia.,Southside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Qld, Australia
| | - Sanjay Warrier
- Department of Breast Surgery, Chris O'Brien Lifehouse, Camperdown, NSW, Australia.,Royal Prince Alfred Institute of Academic Surgery, Sydney University, Sydney, NSW, Australia
| | - Alistair Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA, Australia.,RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Joseph Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia.,UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW, Australia
| | - Sandra O'Toole
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Australian Clinical Laboratories, Northern Beaches Hospital, Frenchs Forest, NSW, Australia
| | - Thomas R Cox
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Elgene Lim
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,St Vincent's Hospital, Darlinghurst, NSW, Australia
| | - X Shirley Liu
- Department of Data Sciences, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander Swarbrick
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
194
|
Yang Y, Yang Y, Yang J, Zhao X, Wei X. Tumor Microenvironment in Ovarian Cancer: Function and Therapeutic Strategy. Front Cell Dev Biol 2020; 8:758. [PMID: 32850861 PMCID: PMC7431690 DOI: 10.3389/fcell.2020.00758] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is one of the leading causes of death in patients with gynecological malignancy. Despite optimal cytoreductive surgery and platinum-based chemotherapy, ovarian cancer disseminates and relapses frequently, with poor prognosis. Hence, it is urgent to find new targeted therapies for ovarian cancer. Recently, the tumor microenvironment has been reported to play a vital role in the tumorigenesis of ovarian cancer, especially with discoveries from genome-, transcriptome- and proteome-wide studies; thus tumor microenvironment may present potential therapeutic target for ovarian cancer. Here, we review the interactions between the tumor microenvironment and ovarian cancer and various therapies targeting the tumor environment.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
195
|
Nazari-Shafti TZ, Neuber S, Garcia Duran A, Xu Z, Beltsios E, Seifert M, Falk V, Stamm C. Human mesenchymal stromal cells and derived extracellular vesicles: Translational strategies to increase their proangiogenic potential for the treatment of cardiovascular disease. Stem Cells Transl Med 2020; 9:1558-1569. [PMID: 32761804 PMCID: PMC7695640 DOI: 10.1002/sctm.19-0432] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) offer great potential for the treatment of cardiovascular diseases (CVDs) such as myocardial infarction and heart failure. Studies have revealed that the efficacy of MSCs is mainly attributed to their capacity to secrete numerous trophic factors that promote angiogenesis, inhibit apoptosis, and modulate the immune response. There is growing evidence that MSC‐derived extracellular vesicles (EVs) containing a cargo of lipids, proteins, metabolites, and RNAs play a key role in this paracrine mechanism. In particular, encapsulated microRNAs have been identified as important positive regulators of angiogenesis in pathological settings of insufficient blood supply to the heart, thus opening a new path for the treatment of CVD. In the present review, we discuss the current knowledge related to the proangiogenic potential of MSCs and MSC‐derived EVs as well as methods to enhance their biological activities for improved cardiac tissue repair. Increasing our understanding of mechanisms supporting angiogenesis will help optimize future approaches to CVD intervention.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ana Garcia Duran
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleftherios Beltsios
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Seifert
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Division of Cardiovascular Surgery, University of Zurich, Zurich, Switzerland
| | - Christof Stamm
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
196
|
Gilard V, Tebani A, Bekri S, Marret S. Intraventricular Hemorrhage in Very Preterm Infants: A Comprehensive Review. J Clin Med 2020; 9:E2447. [PMID: 32751801 PMCID: PMC7465819 DOI: 10.3390/jcm9082447] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/20/2020] [Accepted: 07/25/2020] [Indexed: 11/30/2022] Open
Abstract
Germinal matrix-intraventricular-intraparenchymal hemorrhage (GMH-IVH-IPH) is a major complication of very preterm births before 32 weeks of gestation (WG). Despite progress in clinical management, its incidence remains high before 27 WG. In addition, severe complications may occur such as post-hemorrhagic hydrocephalus and/or periventricular intraparenchymal hemorrhage. IVH is strongly associated with subsequent neurodevelopmental disabilities. For this review, an automated literature search and a clustering approach were applied to allow efficient filtering as well as topic clusters identification. We used a programmatic literature search for research articles related to intraventricular hemorrhage in preterms that were published between January 1990 and February 2020. Two queries ((Intraventricular hemorrhage) AND (preterm)) were used in PubMed. This search resulted in 1093 articles. The data manual curation left 368 documents that formed 12 clusters. The presentation and discussion of the clusters provide a comprehensive overview of existing data on the pathogenesis, complications, neuroprotection and biomarkers of GMH-IVH-IPH in very preterm infants. Clinicians should consider that the GMH-IVH-IPH pathogenesis is mainly due to developmental immaturity of the germinal matrix and cerebral autoregulation impairment. New multiomics investigations of intraventricular hemorrhage could foster the development of predictive biomarkers for the benefit of very preterm newborns.
Collapse
Affiliation(s)
- Vianney Gilard
- Department of Pediatric Neurosurgery, Rouen University Hospital, 76000 Rouen, France;
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
| | - Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France;
| | - Stéphane Marret
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France;
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76000 Rouen, France
| |
Collapse
|
197
|
Wei J, Zhao Y. MiR-185-5p Protects Against Angiogenesis in Polycystic Ovary Syndrome by Targeting VEGFA. Front Pharmacol 2020; 11:1030. [PMID: 32760272 PMCID: PMC7373746 DOI: 10.3389/fphar.2020.01030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/24/2020] [Indexed: 01/08/2023] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is a heterogeneous endocrine disease with high incidences in women of reproductive age. Although miR-185-5p (miR-185) was decreased in PCOS patients, the exact function of miR-185 on PCOS development still requires further investigation. In this study, rat injected with dehydroepiandrosterone (DHEA) was established as a PCOS model. A lentivirus carrying miR-185 was employed to examine its effect on PCOS symptoms. Then we performed the luciferase reporter assay to validate the interactions between miR-185 and vascular endothelial growth factor A (VEGFA). Finally, human ovarian microvascular endothelial cells (HOMECs) were induced by VEGF to explore the role of miR-185 in the angiogenic process. The results showed that miR-185 overexpression improved insulin level alteration and ovarian histological lesion in PCOS rats. We also found that miR-185 reduced the excessive angiogenesis as indicated by alterations of VEGFA, ANGPT1/2, PDGFB/D, α-SMA and CD31 in the ovary of PCOS rats. Luciferase reporter assay identified that VEGFA directly interacted with miR-185, and its expression level was negatively regulated by miR-185. The in vitro results further demonstrated that miR-185-induced suppression of cell proliferation, migration and tube formation was attenuated by VEGF in HOMECs. In summary, this is the first study to show that miR-185 can target VEGFA to inhibit angiogenesis, thus improving the development of PCOS. These findings develop a molecular candidate for PCOS prevention and therapy.
Collapse
Affiliation(s)
- Jingzan Wei
- Department of Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
198
|
Rieger J, Kaessmeyer S, Al Masri S, Hünigen H, Plendl J. Endothelial cells and angiogenesis in the horse in health and disease-A review. Anat Histol Embryol 2020; 49:656-678. [PMID: 32639627 DOI: 10.1111/ahe.12588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/04/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022]
Abstract
The cardiovascular system is the first functional organ in the embryo, and its blood vessels form a widespread conductive network within the organism. Blood vessels develop de novo, by the differentiation of endothelial progenitor cells (vasculogenesis) or by angiogenesis, which is the formation of new blood vessels from existing ones. This review presents an overview of the current knowledge on physiological and pathological angiogenesis in the horse including studies on equine endothelial cells. Principal study fields in equine angiogenesis research were identified: equine endothelial progenitor cells; equine endothelial cells and angiogenesis (heterogeneity, markers and assessment); endothelial regulatory molecules in equine angiogenesis; angiogenesis research in equine reproduction (ovary, uterus, placenta and conceptus, testis); angiogenesis research in pathological conditions (tumours, ocular pathologies, equine wound healing, musculoskeletal system and laminitis). The review also includes a table that summarizes in vitro studies on equine endothelial cells, either describing the isolation procedure or using previously isolated endothelial cells. A particular challenge of the review was that results published are fragmentary and sometimes even contradictory, raising more questions than they answer. In conclusion, angiogenesis is a major factor in several diseases frequently occurring in horses, but relatively few studies focus on angiogenesis in the horse. The challenge for the future is therefore to continue exploring new therapeutic angiogenesis strategies for horses to fill in the missing pieces of the puzzle.
Collapse
Affiliation(s)
- Juliane Rieger
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Sabine Kaessmeyer
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Salah Al Masri
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Hana Hünigen
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Johanna Plendl
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
199
|
Hyun DH. Insights into the New Cancer Therapy through Redox Homeostasis and Metabolic Shifts. Cancers (Basel) 2020; 12:cancers12071822. [PMID: 32645959 PMCID: PMC7408991 DOI: 10.3390/cancers12071822] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Modest levels of reactive oxygen species (ROS) are necessary for intracellular signaling, cell division, and enzyme activation. These ROS are later eliminated by the body’s antioxidant defense system. High amounts of ROS cause carcinogenesis by altering the signaling pathways associated with metabolism, proliferation, metastasis, and cell survival. Cancer cells exhibit enhanced ATP production and high ROS levels, which allow them to maintain elevated proliferation through metabolic reprograming. In order to prevent further ROS generation, cancer cells rely on more glycolysis to produce ATP and on the pentose phosphate pathway to provide NADPH. Pro-oxidant therapy can induce more ROS generation beyond the physiologic thresholds in cancer cells. Alternatively, antioxidant therapy can protect normal cells by activating cell survival signaling cascades, such as the nuclear factor erythroid 2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) pathway, in response to radio- and chemotherapeutic drugs. Nrf2 is a key regulator that protects cells from oxidative stress. Under normal conditions, Nrf2 is tightly bound to Keap1 and is ubiquitinated and degraded by the proteasome. However, under oxidative stress, or when treated with Nrf2 activators, Nrf2 is liberated from the Nrf2-Keap1 complex, translocated into the nucleus, and bound to the antioxidant response element in association with other factors. This cascade results in the expression of detoxifying enzymes, including NADH-quinone oxidoreductase 1 (NQO1) and heme oxygenase 1. NQO1 and cytochrome b5 reductase can neutralize ROS in the plasma membrane and induce a high NAD+/NADH ratio, which then activates SIRT1 and mitochondrial bioenergetics. NQO1 can also stabilize the tumor suppressor p53. Given their roles in cancer pathogenesis, redox homeostasis and the metabolic shift from glycolysis to oxidative phosphorylation (through activation of Nrf2 and NQO1) seem to be good targets for cancer therapy. Therefore, Nrf2 modulation and NQO1 stimulation could be important therapeutic targets for cancer prevention and treatment.
Collapse
Affiliation(s)
- Dong-Hoon Hyun
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
200
|
Montemagno C, Pagès G. Resistance to Anti-angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs. Front Cell Dev Biol 2020; 8:584. [PMID: 32775327 PMCID: PMC7381352 DOI: 10.3389/fcell.2020.00584] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from preexisting one, represents a critical process for oxygen and nutrient supply to proliferating cells, therefore promoting tumor growth and metastasis. The Vascular Endothelial Growth Factor (VEGF) pathway is one of the key mediators of angiogenesis in cancer. Therefore, several therapies including monoclonal antibodies or tyrosine kinase inhibitors target this axis. Although preclinical studies demonstrated strong antitumor activity, clinical studies were disappointing. Antiangiogenic drugs, used to treat metastatic patients suffering of different types of cancers, prolonged survival to different extents but are not curative. In this review, we focused on different mechanisms involved in resistance to antiangiogenic therapies from early stage resistance involving mainly tumor cells to late stages related to the adaptation of the microenvironment.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|