101
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Elucidating Antiangiogenic Potential of Rauwolfia serpentina: VEGFR-2 Targeting-Based Molecular Docking Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6224666. [PMID: 35198035 PMCID: PMC8860507 DOI: 10.1155/2022/6224666] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis plays a critical role in tumorigenesis as it provides the necessary blood supply to the newly grown solid tumor. It helps maintain the tumor microenvironment, promotes tumor development, progression, and metastasis. The vascular epithelial growth factor (VEGF), interacting with the tyrosine kinase receptor VEGFR-2 on endothelial cells, exerts its proangiogenic activity. Hence, targeting the VEGFR-2 signaling is considered a promising strategy to inhibit angiogenesis and thus cancer treatment. This study aims to identify the bioactive compounds derived from the medicinal herb Rauwolfia serpentina that effectively binds with VEGFR-2. The bioactive compounds of R. serpentina were first screened for their physicochemical properties using the DataWarrior program (version 5.5.0). Finally, 17 compounds that obeyed Lipinski's rule of five and showed good drug-likeness were selected for molecular docking studies. Molecular docking results showed that the ligands ajmalicidine, 1, 2-dihydrovomilenine, rauwolscine, yohimbine, ajmaline, and papaverine interact strongly with the target VEGFR-2 receptor. Hydrogen bonds and hydrophobic interactions stabilized the interactions of these compounds with VEGFR-2. These compounds showed favourable drug-like properties and possess no significant toxicity. Therefore, the findings of this study indicate that the compounds derived from R. serpentina can be considered for the development of antiangiogenic drug candidates by targeting VEGFR-2.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
102
|
Fu L, Deng R, Huang Y, Yang X, Jiang N, Zhou J, Lin C, Chen S, Wu L, Cui Q, Yun J. DGKA interacts with SRC/FAK to promote the metastasis of non-small cell lung cancer. Cancer Lett 2022; 532:215585. [PMID: 35131384 DOI: 10.1016/j.canlet.2022.215585] [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: 11/15/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/25/2022]
Abstract
Metastasis is responsible for the high mortality rate of lung cancer, but its underlying molecular mechanisms are poorly understood. Here, we demonstrated that the expression of diacylglycerol kinase alpha (DGKA) was elevated in the metastatic lesions of non-small cell lung cancer (NSCLC) and correlated with poor survival. Mechanistic studies revealed a direct physical interaction as well as a mutual regulation among DGKA, proto-oncogene tyrosine-protein kinase Src (SRC), and focal adhesion kinase 1 (FAK) proteins. The C-terminal domain of DGKA was responsible for the SRC SH3 domain binding, while the catalytic domain of DGKA interacted with the FREM domain of FAK. DGKA phosphorylated the SRC protein at Tyr416 and the FAK protein at Tyr397 to form and activate the DGKA/SRC/FAK complex, thus initiating the downstream WNT/β-catenin and VEGF signaling pathways, promoting epithelial-mesenchymal transition (EMT) and angiogenesis, and resulting in the metastasis of NSCLC. DGKA knockdown inhibited the invasive phenotype of NSCLC cells in vitro. Pharmacologic ablation of DGKA inhibited the metastasis of NSCLC cells in vivo, and this was reversed by the overexpression of DGKA. These results suggested that DGKA was a potential prognostic biomarker as well as a promising therapeutic target for NSCLC, especially when there was lymphatic or distant metastasis.
Collapse
Affiliation(s)
- Lingyi Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Ru Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Yuhua Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Neng Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jing Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Censhan Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Shilu Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Liyan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Qian Cui
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
103
|
Rosano S, Parab S, Noghero A, Corà D, Bussolino F. Long Non-Coding RNA LINC02802 Regulates In Vitro Sprouting Angiogenesis by Sponging microRNA-486-5p. Int J Mol Sci 2022; 23:ijms23031653. [PMID: 35163581 PMCID: PMC8836176 DOI: 10.3390/ijms23031653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
In the last several years, accumulating evidence indicates that noncoding RNAs, especially long-noncoding RNAs (lncRNAs) and microRNAs, play essential roles in regulating angiogenesis. However, the contribution of lncRNA-mediated competing-endogenous RNA (ceRNA) activity in the control of capillary sprouting from the pre-existing ones has not been described so far. Here, by exploiting the transcriptomic profile of VEGF-A-activated endothelial cells in a consolidate three-dimensional culture system, we identified a list of lncRNAs whose expression was modified during the sprouting process. By crossing the lncRNAs with a higher expression level and the highest fold change value between unstimulated and VEGF-A-stimulated endothelial cells, we identified the unknown LINC02802 as the best candidate to take part in sprouting regulation. LINC02802 was upregulated after VEGF-A stimulation and its knockdown resulted in a significant reduction in sprouting activity. Mechanistically, we demonstrated that LINC02802 acts as a ceRNA in the post-transcriptional regulation of Mastermind-like-3 (MAML3) gene expression through a competitive binding with miR-486-5p. Taken together, these results suggest that LINC02802 plays a critical role in preventing the miR-486-5p anti-angiogenic effect and that this inhibitory effect results from the reduction in MAML3 expression.
Collapse
Affiliation(s)
- Stefania Rosano
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
- Correspondence:
| | - Sushant Parab
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
| | - Alessio Noghero
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA;
| | - Davide Corà
- Department of Translational Medicine, Piemonte Orientale University, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Diseases—CAAD, 28100 Novara, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
| |
Collapse
|
104
|
Alam M, Ali S, Ashraf GM, Bilgrami AL, Yadav DK, Hassan MI. Epigallocatechin 3-gallate: From green tea to cancer therapeutics. Food Chem 2022; 379:132135. [PMID: 35063850 DOI: 10.1016/j.foodchem.2022.132135] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 01/09/2022] [Indexed: 12/13/2022]
Abstract
Epigallocatechin 3-gallate (EGCG) possesses various biological functions, including anti-cancer and anti-inflammatory properties. EGCG is an abundant polyphenolic component originating from green tea extract that has exhibited versatile bioactivities in combating several cancers. This review highlights the pharmacological features of EGCG and its therapeutic implications in cancer and other metabolic diseases. It modulates numerous signaling pathways, regulating cells' undesired survival and proliferation, thus imparting strong tumor chemopreventive and therapeutic effects. EGCG initiates cell death through the intrinsic pathway and causes inhibition of EGFR, STAT3, and ERK pathways in several cancers. EGCG alters and inhibits ERK1/2, NF-κB, and Akt-mediated signaling, altering the Bcl-2 family proteins ratio and activating caspases in tumor cells. This review focuses on anti-cancer, anti-oxidant, anti-inflammatory, anti-angiogenesis, and apoptotic effects of EGCG. We further highlighted the potential of EGCG in different types of cancer, emphasizing clinical trials formulations that further improve our understanding of the therapeutic management of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar L Bilgrami
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon City 21924, South Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
105
|
Manzoor S, Wani SM, Mir SA, Rizwan D. Role of probiotics and prebiotics in mitigation of different diseases. Nutrition 2022; 96:111602. [PMID: 35182833 DOI: 10.1016/j.nut.2022.111602] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022]
|
106
|
Abstract
Based on the analysis of published data, the review provides information on the role and mechanisms of angiogenesis in the development of eye diseases. It has been shown that the developing inflammatory process associated with infections or damage to the organ of vision almost always leads to the appearance of newly formed vessels in the avascular cornea. The progression, in particular, of age-related macular degeneration is associated with the immune-mediated development of angiogenesis processes. A key inducer of angiogenesis is vascular endothelial growth factor (VEGF), whose activity can be enhanced by a number of pro-inflammatory cytokines (tumor necrosis factor alpha, TNF-), growth (fibroblast growth factor, FGF) and transforming factors (transforming growth factor beta, TGF- ). In addition, VEGF overproduction is mediated by an imbalance of pro-angiogenic (angiogenin) and anti-angiogenic (angiostatin, vasostatin, endostatin; tissue inhibitors of matrix metalloproteinases) factors. Antiangiogenic activity based on inhibition of vascular endothelial growth factor (VEGF) has been successfully used in the treatment of a number of eye diseases, such as exudative age-related macular degeneration and diabetic macular edema, the pathogenesis of which is based on the growth of newly formed vessels. The review presents information on the main anti-angiogenic drugs for intravitreal administration, used in ophthalmology.
Collapse
|
107
|
Li Y, Zhang K, Yang F, Jiao D, Li M, Zhao X, Xu C, Liu S, Li H, Shi S, Yang B, Yang L, Han D, Wen W, Qin W. Prognostic Value of Vascular-Expressed PSMA and CD248 in Urothelial Carcinoma of the Bladder. Front Oncol 2021; 11:771036. [PMID: 34869004 PMCID: PMC8635966 DOI: 10.3389/fonc.2021.771036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/27/2021] [Indexed: 11/22/2022] Open
Abstract
Background Urothelial carcinoma of the bladder (UCB) is a common cancer of the urinary system. Despite substantial improvements in available treatment options, the survival outcome of patients with advanced UCB is unsatisfactory. Therefore, it is necessary to identify new prognostic biomarkers for monitoring and therapy guidance of UCB. In recent years, prostate-specific membrane antigen (PSMA) and CD248 have been identified promising candidate bio7markers. Methods In this study, we first examined PSMA and CD248 expression in tissues from 124 patients with UCB using immunohistochemical and immunofluorescent staining. We then analyzed the association between the expression of the two biomarkers and other clinicopathological features and prognosis. Finally, we performed bioinformatic analysis of CD248 and FOLH 1 (PSMA) using the TCGA-BLCA dataset to explore the underlying mechanism of PSMA and CD248 in the progression of UCB. Results Among the 124 cases, PSMA and CD248 were confirmed to be expressed in tumor-associated vessels. Vascular PSMA and CD248 expression levels were associated significantly with several deteriorated clinicopathological features. Furthermore, using univariate and multivariate Cox analyses, high vascular PSMA and CD248 expression levels were observed to be associated significantly with poor prognosis in patients with UCB. As risk factors, both PSMA and CD248 expression showed good performance to predict prognosis. Furthermore, combining these vascular molecules with other clinical risk factors generated a risk score that could promote predictive performance. Bioinformatic analysis showed that both PSMA and CD248 might contribute to angiogenesis and promote further progression of UCB. Conclusion Both PSMA and CD248 are specifically expressed in the tumor-associated vasculature of UCB. These two molecules might be used as novel prognostic biomarkers and vascular therapeutic targets for UCB.
Collapse
Affiliation(s)
- Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dian Jiao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mingyang Li
- Department of Pathology, Fourth Military Medical University, Xi'an, China
| | - Xiaolong Zhao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongji Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shengjia Shi
- Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, China
| | - Bo Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lijun Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
108
|
Becker-Greene D, Li H, Perez-Cremades D, Wu W, Bestepe F, Ozdemir D, Niosi CE, Aydogan C, Orgill DP, Feinberg MW, Icli B. MiR-409-3p targets a MAP4K3-ZEB1-PLGF signaling axis and controls brown adipose tissue angiogenesis and insulin resistance. Cell Mol Life Sci 2021; 78:7663-7679. [PMID: 34698882 PMCID: PMC8655847 DOI: 10.1007/s00018-021-03960-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/09/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
Endothelial cells (ECs) within the microvasculature of brown adipose tissue (BAT) are important in regulating the plasticity of adipocytes in response to increased metabolic demand by modulating the angiogenic response. However, the mechanism of EC-adipocyte crosstalk during this process is not completely understood. We used RNA sequencing to profile microRNAs derived from BAT ECs of obese mice and identified an anti-angiogenic microRNA, miR-409-3p. MiR-409-3p overexpression inhibited EC angiogenic properties; whereas, its inhibition had the opposite effects. Mechanistic studies revealed that miR-409-3p targets ZEB1 and MAP4K3. Knockdown of ZEB1/MAP4K3 phenocopied the angiogenic effects of miR-409-3p. Adipocytes co-cultured with conditioned media from ECs deficient in miR-409-3p showed increased expression of BAT markers, UCP1 and CIDEA. We identified a pro-angiogenic growth factor, placental growth factor (PLGF), released from ECs in response to miR-409-3p inhibition. Deficiency of ZEB1 or MAP4K3 blocked the release of PLGF from ECs and PLGF stimulation of 3T3-L1 adipocytes increased UCP1 expression in a miR-409-3p dependent manner. MiR-409-3p neutralization improved BAT angiogenesis, glucose and insulin tolerance, and energy expenditure in mice with diet-induced obesity. These findings establish miR-409-3p as a critical regulator of EC-BAT crosstalk by modulating a ZEB1-MAP4K3-PLGF signaling axis, providing new insights for therapeutic intervention in obesity.
Collapse
Affiliation(s)
- Dakota Becker-Greene
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Hao Li
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Daniel Perez-Cremades
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Department of Physiology, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Winona Wu
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Furkan Bestepe
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA
| | - Denizhan Ozdemir
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Carolyn E Niosi
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA
| | - Ceren Aydogan
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Dennis P Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA.
| | - Basak Icli
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA.
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
109
|
Inhibition of Vascular Smooth Muscle and Cancer Cell Proliferation by New VEGFR Inhibitors and Their Immunomodulator Effect: Design, Synthesis, and Biological Evaluation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8321400. [PMID: 34745424 PMCID: PMC8568530 DOI: 10.1155/2021/8321400] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/16/2021] [Indexed: 11/25/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation has an important role in the pathogenesis of both atherosclerosis restenosis and hypertension. Vascular endothelial growth factor (VEGF) has been shown to stimulate VSMC proliferation. In addition, angiogenesis is one of the hallmarks of cancerous growth. VEGF is the key modulator for the initial stages of angiogenesis that acts through the endothelial-specific receptor tyrosine kinases (VEGFRs). VEGFR-2 blockage is a good approach for suppression of angiogenesis. In order to discover novel VEGFR-2 TK inhibitors, we have designed and synthesized three new series of pyridine-containing compounds. The new compounds were all screened against a panel of three cell lines (HepG-2, HCT-116, and MCF-7). Promising results encouraged us to additionally evaluate the most active members for their in vitro VEGFR-2 inhibitory effect. Compound 7a, which is the most potent candidate, revealed a significant increase in caspase-3 level by 7.80-fold when compared to the control. In addition, Bax and Bcl-2 concentration levels showed an increase in the proapoptotic protein Bax (261.4 Pg/ml) and a decrease of the antiapoptotic protein Bcl-2 (1.25 Pg/ml) compared to the untreated cells. Furthermore, compound 7a arrested the cell cycle in the G2/M phase with induction of apoptosis. The immunomodulatory effect of compound 7a, the most active member, showed a reduction in TNF-α by 87%. Also, compound 7a caused a potent inhibitory effect on smooth muscle proliferation. Docking studies were also performed to get better insights into the possible binding mode of the target compounds with VEGFR-2 active sites.
Collapse
|
110
|
Qian Y, Li Y, Li R, Yang T, Jia R, Ge YZ. circ-ZNF609: A potent circRNA in human cancers. J Cell Mol Med 2021; 25:10349-10361. [PMID: 34697887 PMCID: PMC8581316 DOI: 10.1111/jcmm.16996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/22/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Circular RNAs (circRNAs) are a novel group of endogenous RNAs with a circular structure. Growing evidence indicates that circRNAs are involved in a variety of human diseases including malignancies. CircRNA ZNF609 (circ‐ZNF609), derived from the ZNF609 gene sequence, has been demonstrated to be involved in the development and progression of many diseases. circ‐ZNF609 is thought to be a viable diagnostic and prognostic biomarker for several diseases and might be a new therapeutic target, but further research is needed to accelerate clinical application. Here, we review the biogenesis and function of circRNAs and the functional roles and molecular mechanism related to circ‐ZNF609 in neoplasms and other diseases.
Collapse
Affiliation(s)
- Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rongfei Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tianli Yang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
111
|
The emerging role of miR-200 family in metastasis: focus on EMT, CSCs, angiogenesis, and anoikis. Mol Biol Rep 2021; 48:6935-6947. [PMID: 34510322 DOI: 10.1007/s11033-021-06666-6] [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: 04/24/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cancer is the second major threat to human society and one of the main challenges facing healthcare systems. One of the main problems of cancer care is the metastases of cancer cells that cause 90% of deaths due to cancer. Multiple molecular mechanisms are involved in cancer cell metastasis. Therefore, a better understanding of these molecular mechanisms is necessary for designing restrictive strategies against cancer cell metastasis. Accumulating data suggests that MicroRNAs (miRNAs) are involved in metastasis and invasion of human tumors through regulating multiple genes expression levels that are involved in molecular mechanisms of metastasis. The goal of this review is to present the molecular pathways by which the miR 200 family manifests its effects on EMT, cancer stem cells, angiogenesis, anoikis, and the effects of tumor cell metastases. METHODS A detailed literature search was conducted to find information about the role of the miR-200 family in the processes involved in metastasis in various databases. RESULTS Numerous lines of evidence revealed an association between the mir-200 family and metastasis of human tumors by impressing processes such as cancer stem cells, EMT, angiogenesis, and anoikis. CONCLUSIONS Understanding the molecular mechanisms associated with metastasis in which the miR-200 family is involved can be effective in treating metastatic cancers.
Collapse
|
112
|
miR-329-containing exosomes derived from breast tumor cells suppress VEGF and KDM1A expression in endothelial cells. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
113
|
de Melo Bisneto AV, Fernandes AS, Velozo Sá VDS, Véras JH, Soares ETS, da Silva Santos AF, Cardoso CG, Silveira-Lacerda EDP, Carneiro CC, Chen-Chen L. Anti-angiogenic activity of azathioprine. Microvasc Res 2021; 138:104234. [PMID: 34478745 DOI: 10.1016/j.mvr.2021.104234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/16/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022]
Abstract
Azathioprine (AZA) is the main drug used in immunomodulatory therapy in post-transplant patients or with autoimmune diseases. However, no study has evaluated the AZA angiogenic response. Therefore, this study investigated the effects of AZA on the angiogenic process through macroscopic, histological, and immunohistochemical analyses in chick embryo chorioallantoic membrane (CAM). Our results showed potent anti-angiogenic activity of AZA at the higher concentrations tested in the CAM assay. The histological analysis of CAM confirmed this effect, since AZA induced a significant reduction in all parameters evaluated. In addition, immunohistochemical evaluation of CAM revealed that AZA decreased TGF-β and VEGF levels, important cytokines involved in the angiogenic process. Therefore, the AZA anti-angiogenic effect identified in our study provides new information for the possible application of this drug in anticancer treatment.
Collapse
Affiliation(s)
- Abel Vieira de Melo Bisneto
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Amanda Silva Fernandes
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Vivianne de Souza Velozo Sá
- Laboratory of Molecular Genetics and Cytogenetics, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Jefferson Hollanda Véras
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Estéfane Thaíne Sodré Soares
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | | | - Clever Gomes Cardoso
- Department of Morphology of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Elisângela de Paula Silveira-Lacerda
- Laboratory of Molecular Genetics and Cytogenetics, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Cristiene Costa Carneiro
- Institute of Health Sciences, Universidade Paulista - Campus Flamboyant, 74845-090 Goiânia, Brazil
| | - Lee Chen-Chen
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil.
| |
Collapse
|
114
|
Marcheteau E, Farge T, Pérès M, Labrousse G, Tenet J, Delmas S, Chusseau M, Duprez-Paumier R, Franchet C, Dalenc F, Imbert C, Noujarède J, Colacios C, Prats H, Cabon F, Ségui B. Thrombospondin-1 Silencing Improves Lymphocyte Infiltration in Tumors and Response to Anti-PD-1 in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:4059. [PMID: 34439212 PMCID: PMC8391594 DOI: 10.3390/cancers13164059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 01/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is notoriously aggressive with a high metastatic potential, and targeted therapies are lacking. Using transcriptomic and histologic analysis of TNBC samples, we found that a high expression of thrombospondin-1 (TSP1), a potent endogenous inhibitor of angiogenesis and an activator of latent transforming growth factor beta (TGF-β), is associated with (i) gene signatures of epithelial-mesenchymal transition and TGF-β signaling, (ii) metastasis and (iii) a reduced survival in TNBC patients. In contrast, in tumors expressing low levels of TSP1, gene signatures of interferon gamma (IFN-γ) signaling and lymphocyte activation were enriched. In TNBC biopsies, TSP1 expression inversely correlated with the CD8+ tumor-infiltrating lymphocytes (TILs) content. In the 4T1 metastatic mouse model of TNBC, TSP1 silencing did not affect primary tumor development but, strikingly, impaired metastasis in immunocompetent but not in immunodeficient nude mice. Moreover, TSP1 knockdown increased tumor vascularization and T lymphocyte infiltration and decreased TGF-β activation in immunocompetent mice. Noteworthy was the finding that TSP1 knockdown increased CD8+ TILs and their programmed cell death 1 (PD-1) expression and sensitized 4T1 tumors to anti-PD-1 therapy. TSP1 inhibition might thus represent an innovative targeted approach to impair TGF-β activation and breast cancer cell metastasis and improve lymphocyte infiltration in tumors, and immunotherapy efficacy in TNBC.
Collapse
Affiliation(s)
- Elie Marcheteau
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- SeleXel, 1 Place Pierre Potier, BP 50624, CEDEX 1, 31106 Toulouse, France; (S.D.); (M.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Michaël Pérès
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
| | - Guillaume Labrousse
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Julie Tenet
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Stéphanie Delmas
- SeleXel, 1 Place Pierre Potier, BP 50624, CEDEX 1, 31106 Toulouse, France; (S.D.); (M.C.)
| | - Maud Chusseau
- SeleXel, 1 Place Pierre Potier, BP 50624, CEDEX 1, 31106 Toulouse, France; (S.D.); (M.C.)
| | - Raphaëlle Duprez-Paumier
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France; (R.D.-P.); (C.F.); (F.D.)
| | - Camille Franchet
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France; (R.D.-P.); (C.F.); (F.D.)
| | - Florence Dalenc
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France; (R.D.-P.); (C.F.); (F.D.)
| | - Caroline Imbert
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
| | - Justine Noujarède
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Céline Colacios
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Hervé Prats
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Florence Cabon
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- SeleXel, 1 Place Pierre Potier, BP 50624, CEDEX 1, 31106 Toulouse, France; (S.D.); (M.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
| | - Bruno Ségui
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, 2 Aavenue Hubert Curien, CEDEX 1, 31047 Toulouse, France; (E.M.); (T.F.); (M.P.); (G.L.); (J.T.); (C.I.); (J.N.); (C.C.); (H.P.); (F.C.)
- Université Toulouse III—Paul Sabatier, 118 Rte de Narbonne, 31062 Toulouse, France
- Equipe Labellisée par la Fondation ARC—Association Pour la Recherche sur le Cancer, 94803 Villejuif, France
| |
Collapse
|
115
|
Oudart JB, Villemin M, Brassart B, Sellier C, Terryn C, Dupont-Deshorgue A, Monboisse JC, Maquart FX, Ramont L, Brassart-Pasco S. F4, a collagen XIX-derived peptide, inhibits tumor angiogenesis through αvβ3 and α5β1 integrin interaction. Cell Adh Migr 2021; 15:215-223. [PMID: 34308743 PMCID: PMC8312610 DOI: 10.1080/19336918.2021.1951425] [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] [Indexed: 11/25/2022] Open
Abstract
We previously demonstrated that F4 peptide (CNPEDCLYPVSHAHQR) from collagen XIX was able to inhibit melanoma cell migrationin vitro and cancer progression in a mouse melanoma model. The aim of the present work was to study the anti-angiogenic properties of F4 peptide. We demonstrated that F4 peptide inhibited VEGF-induced pseudo-tube formation on Matrigel by endothelial cells and endothelial sprouting in a rat aortic ring assay. By affinity chromatography, we identified αvβ3 and α5β1 integrins as potential receptors for F4 peptide on endothelial cell surface. Using solid phase assays, we proved the direct interaction between F4 and both integrins. Taken together, our results demonstrate that F4 peptide is a potent antitumor agent inhibiting both angiogenesis and tumor cell migration.
Collapse
Affiliation(s)
- Jean-Baptiste Oudart
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Matthieu Villemin
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Bertrand Brassart
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Christèle Sellier
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Christine Terryn
- PICT, Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Aurélie Dupont-Deshorgue
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Jean Claude Monboisse
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - François-Xavier Maquart
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Laurent Ramont
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Sylvie Brassart-Pasco
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), Reims, France
| |
Collapse
|
116
|
Raica M, Cimpean AM, Ferician OC, Ferician AM. Tumour-associated Angiogenesis and Intermediate Blood Vessels in Renal Cell Carcinoma. CANCER DIAGNOSIS & PROGNOSIS 2021; 1:231-234. [PMID: 35399306 PMCID: PMC8962796 DOI: 10.21873/cdp.10031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND/AIM Renal cell carcinoma is strongly vascularized, and formation of new blood vessels is a complex and multi-step process. In this study, we analysed the subtypes of intermediate blood vessels, as shown by double immunohistochemistry. MATERIALS AND METHODS Tumour-associated blood vessels were identified by double immunostaining based on CD34 and smooth muscle cell actin. Blood vessels were classified both quantitatively and qualitatively based on the expression of the aforementioned two markers. The main criteria to sub-classify intermediate blood vessels was the presence, distribution, and arrangement of perivascular cells. RESULTS We described three subtypes of intermediate blood vessels found particularly in the tumour area: Subtype 1 lacked perivascular cells, subtype 2 showed scattered pericytes attached to the vascular wall, and subtype 3 showed a continuous layer of perivascular cells on one side. CONCLUSION We describe for the first time three subtypes of renal cell carcinoma-associated intermediate blood vessels, which could be important in prognosis and as potential targets for anti-vascular therapy.
Collapse
Affiliation(s)
- Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center,Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center,Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | | | - Adela Maria Ferician
- Department of Urology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
117
|
Marzo T, La Mendola D. The Effects on Angiogenesis of Relevant Inorganic Chemotherapeutics. Curr Top Med Chem 2021; 21:73-86. [PMID: 33243124 DOI: 10.2174/1568026620666201126163436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a key process allowing the formation of blood vessels. It is crucial for all the tissues and organs, ensuring their function and growth. Angiogenesis is finely controlled by several mechanisms involving complex interactions between pro- or antiangiogenic factors, and an imbalance in this control chain may result in pathological conditions. Metals as copper, zinc and iron cover an essential role in regulating angiogenesis, thus therapies having physiological metals as target have been proposed. In addition, some complexes of heavier metal ions (e.g., Pt, Au, Ru) are currently used as established or experimental anticancer agents targeting genomic or non-genomic targets. These molecules may affect the angiogenic mechanisms determining different effects that have been only poorly and non-systematically investigated so far. Accordingly, in this review article, we aim to recapitulate the impact on the angiogenic process of some reference anticancer drugs, and how it is connected to the overall pharmacological effects. In addition, we highlight how the activity of these drugs can be related to the role of biological essential metal ions. Overall, this may allow a deeper description and understanding of the antineoplastic activity of both approved or experimental metal complexes, providing important insights for the synthesis of new inorganic drugs able to overcome resistance and recurrence phenomena.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
118
|
Andleeb F, Katta N, Gruslova A, Muralidharan B, Estrada A, McElroy AB, Ullah H, Brenner AJ, Milner TE. Differentiation of Brain Tumor Microvasculature From Normal Vessels Using Optical Coherence Angiography. Lasers Surg Med 2021; 53:1386-1394. [PMID: 34130353 DOI: 10.1002/lsm.23446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND OBJECTIVES Despite rapid advances and discoveries in medical imaging, monitoring therapeutic efficacy for malignant gliomas and monitoring tumor vasculature remains problematic. The purpose of this study is to utilize optical coherence angiography for vasculature characterization inside and surrounding brain tumors in a murine xenograft brain tumor model. Features included in our analysis include fractional blood volume, vessel tortuosity, diameter, orientation, and directionality. STUDY DESIGN/MATERIALS AND METHODS In this study, five tumorous mice models at 4 weeks of age were imaged. Human glioblastoma cells were injected into the brain and allowed to grow for 4 weeks and then imaged using optical coherence tomography. RESULTS Results suggest that blood vessels outside the tumor contain a greater fractional blood volume as compared with vessels inside the tumor. Vessels inside the tumor are more tortuous as compared with those outside the tumor. Results indicate that vessels near the tumor margin are directed inward towards the tumor while normal vessels show a more random orientation. CONCLUSION Quantification of vascular microenvironments in brain gliomas can provide functional vascular parameters to aid various diagnostic and therapeutic studies. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Farah Andleeb
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA.,Biophotonics Research Lab, Institute of Physics, The Islamia University, Bahawalpur, Bahawalpur, Punjab, 63100, Pakistan.,Department of Physics, Government Sadiq College Women University Bahawalpur, Bahwalpur, Punjab, 63100, Pakistan
| | - Nitesh Katta
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA
| | - Aleksandra Gruslova
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
| | - Bharadwaj Muralidharan
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA
| | - Arnold Estrada
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA
| | - Austin B McElroy
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA
| | - Hafeez Ullah
- Biophotonics Research Lab, Institute of Physics, The Islamia University, Bahawalpur, Bahawalpur, Punjab, 63100, Pakistan
| | - Andrew J Brenner
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
| | - Thomas E Milner
- Department of Biomedical Engineering, The University of Texas Austin, Austin, Texas, 78712, USA
| |
Collapse
|
119
|
Sameri S, Mohammadi C, Mehrabani M, Najafi R. Targeting the hallmarks of cancer: the effects of silibinin on proliferation, cell death, angiogenesis, and migration in colorectal cancer. BMC Complement Med Ther 2021; 21:160. [PMID: 34059044 PMCID: PMC8168007 DOI: 10.1186/s12906-021-03330-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
Background Silibinin, as a chemopreventive agent, has shown anti-cancer efficacy against different types of cancers. In the present study, we investigated the anti-cancer activities of silibinin on CT26 mouse colon cell line. Methods CT26 cells were treated with different concentrations of silibinin. To examine the cytotoxic effect of silibinin on proliferation, apoptosis, autophagy, angiogenesis, and migration, MTT, colony-forming assay, Annexin V/PI flow cytometry, RT-qPCR, and Scratch assay were used. Results Silibinin was found to significantly reduce CT26 cells survival. Furthermore, silibinin strongly induced apoptosis and autophagy by up-regulating the expression of Bax, Caspase-3, Atg5, Atg7 and BECN1 and down-regulating Bcl-2. Silibinin considerably down-regulated the expression of COX-2, HIF-1α, VEGF, Ang-2, and Ang-4 as well as the expression of MMP-2, MMP-9, CCR-2 and CXCR-4. Conclusions The present study revealed that silibinin shows anticancer activities by targeting proliferation, cell survival, angiogenesis, and migration of CT26 cells.
Collapse
Affiliation(s)
- Saba Sameri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Chiman Mohammadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
120
|
Shiraki A, Sakimoto S, Eguchi M, Kanai M, Hara C, Fukushima Y, Nishida K, Kawasaki R, Sakaguchi H, Nishida K. Analysis of Progressive Neovascularization in Diabetic Retinopathy Using Widefield OCT Angiography. Ophthalmol Retina 2021; 6:153-160. [PMID: 34051418 DOI: 10.1016/j.oret.2021.05.011] [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: 12/12/2020] [Revised: 04/27/2021] [Accepted: 05/13/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE To document enlarged neovascularization elsewhere (NVE) quantitatively and morphologically using widefield swept-source (SS) OCT angiography (OCTA) with vitreoretinal interface (VRI) slab images. DESIGN Retrospective, observational imaging study. PARTICIPANTS The study included 46 NVE examples in 25 eyes of 21 consecutive patients who demonstrated severe proliferative diabetic retinopathy with NVE between March 2018 and June 2020 at Osaka University Hospital. METHODS All patients underwent ophthalmologic examination, including ultra-widefield fluorescein angiography and widefield SS OCTA scans. MAIN OUTCOME MEASURES We evaluated the area and the vascular density (VD) of NVE lesions detected on five 12 × 12-mm2 or two 15 × 9-mm2 SS OCTA panoramic VRI slab images obtained at the first and final visits. RESULTS At baseline, the mean NVE area on OCTA was 1.85 ± 2.81 mm2, and the VD of the NVE lesions was 73.9 ± 14.6%. At the final visit, the mean NVE area on OCTA was 2.14 ± 3.14 mm2, and the mean VD of the NVE lesions was 65.3 ± 17.1%. The average NVE size change (square millimeters per month) was associated significantly with the ischemic index (P = 0.009). Growth of NVE area was classified into 2 patterns: round (61.8%) and ramified (38.2%). The round group tended to have a larger ischemic index at baseline than the ramified group (P = 0.0375). CONCLUSIONS We quantified the size and density of NVE lesions over time. The NVE size increase was associated significantly with the severity of ischemic changes. Furthermore, the round growth pattern was correlated significantly with the ischemic index. These findings suggest that the morphologic features of NVE are associated with more severe ischemia.
Collapse
Affiliation(s)
- Akihiko Shiraki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Susumu Sakimoto
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan.
| | - Mami Eguchi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masanori Kanai
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chikako Hara
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoko Fukushima
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Kentaro Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryo Kawasaki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hirokazu Sakaguchi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
121
|
Gutiérrez LM, Valenzuela Alvarez M, Yang Y, Spinelli F, Cantero MJ, Alaniz L, García MG, Kleinerman ES, Correa A, Bolontrade MF. Up-regulation of pro-angiogenic molecules and events does not relate with an angiogenic switch in metastatic osteosarcoma cells but to cell survival features. Apoptosis 2021; 26:447-459. [PMID: 34024019 DOI: 10.1007/s10495-021-01677-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2021] [Indexed: 01/22/2023]
Abstract
Osteosarcoma (OS) is the most frequent malignant bone tumor, affecting predominantly children. Metastases represent a major clinical challenge and an estimated 80% would present undetectable micrometastases at diagnosis. The identification of metastatic traits and molecules would impact in micrometastasis management. We demonstrated that OS LM7 metastatic cells secretome was able to induce microvascular endothelium cell rearrangements, an angiogenic-related trait. A proteomic analysis indicated a gain in angiogenic-related pathways in these cells, as compared to their parental-non-metastatic OS SAOS2 cells counterpart. Further, factors with proangiogenic functions like VEGF and PDGF were upregulated in LM7 cells. However, no differential angiogenic response was induced by LM7 cells in vivo. Regulation of the Fas-FasL axis is key for OS cells to colonize the lungs in this model. Analysis of the proteomic data with emphasis in apoptosis pathways and related processes revealed that the percentage of genes associated with those, presented similar levels in SAOS2 and LM7 cells. Further, the balance of expression levels of proteins with pro- and antiapoptotic functions in both cell types was subtle. Interestingly and of relevance to the model, Fas associated Factor 1 (FAF1), which participates in Fas signaling, was present in LM7 cells and was not detected in SAOS2 cells. The subtle differences in apoptosis-related events and molecules, together with the reported cell-survival functions of the identified angiogenic factors and the increased survival features that we observed in LM7 cells, suggest that the gain in angiogenesis-related pathways in metastatic OS cells would relate to a prosurvival switch rather to an angiogenic switch as an advantage feature to colonize the lungs. OS metastatic cells also displayed higher adhesion towards microvascular endothelium cells suggesting an advantage for tissue colonization. A gain in angiogenesis pathways and molecules does not result in major angiogenic potential. Together, our results suggest that metastatic OS cells would elicit signaling associated to a prosurvival phenotype, allowing homing into the hostile site for metastasis. During the gain of metastatic traits process, cell populations displaying higher adhesive ability to microvascular endothelium, negative regulation of the Fas-FasL axis in the lung parenchyma and a prosurvival switch, would be selected. This opens a new scenario where antiangiogenic treatments would affect cell survival rather than angiogenesis, and provides a molecular panel of expression that may help in distinguishing OS cells with different metastatic potential.
Collapse
Affiliation(s)
- Luciana M Gutiérrez
- Remodeling Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), Potosí 4240, C1199ACL, CABA, Argentina
| | - Matías Valenzuela Alvarez
- Remodeling Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), Potosí 4240, C1199ACL, CABA, Argentina
| | - Yuanzheng Yang
- Division of Pediatrics and Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit #853, Houston, TX, 77030, USA
| | | | - María José Cantero
- Facultad de Ciencias Biomédicas, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET, Universidad Austral, Pilar, Buenos Aires, Argentina
| | - Laura Alaniz
- CITNOBA CONICET-UNNOBA, Jorge Newbery 261, B6000, Junín, Argentina
| | - Mariana G García
- Facultad de Ciencias Biomédicas, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET, Universidad Austral, Pilar, Buenos Aires, Argentina
| | - Eugenie S Kleinerman
- Division of Pediatrics and Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit #853, Houston, TX, 77030, USA
| | | | - Marcela F Bolontrade
- Remodeling Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), Potosí 4240, C1199ACL, CABA, Argentina.
| |
Collapse
|
122
|
Cappelli HC, Guarino BD, Kanugula AK, Adapala RK, Perera V, Smith MA, Paruchuri S, Thodeti CK. Transient receptor potential vanilloid 4 channel deletion regulates pathological but not developmental retinal angiogenesis. J Cell Physiol 2021; 236:3770-3779. [PMID: 33078410 PMCID: PMC7920906 DOI: 10.1002/jcp.30116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels are mechanosensitive ion channels that regulate systemic endothelial cell (EC) functions such as vasodilation, permeability, and angiogenesis. TRPV4 is expressed in retinal ganglion cells, Müller glia, pigment epithelium, microvascular ECs, and modulates cell volume regulation, calcium homeostasis, and survival. TRPV4-mediated physiological or pathological retinal angiogenesis remains poorly understood. Here, we demonstrate that TRPV4 is expressed, functional, and mechanosensitive in retinal ECs. The genetic deletion of TRPV4 did not affect postnatal developmental angiogenesis but increased pathological neovascularization in response to oxygen-induced retinopathy (OIR). Retinal vessels from TRPV4 knockout mice subjected to OIR exhibited neovascular tufts that projected into the vitreous humor and displayed reduced pericyte coverage compared with wild-type mice. These results suggest that TRPV4 is a regulator of retinal angiogenesis, its deletion augments pathological retinal angiogenesis, and that TRPV4 could be a novel target for the development of therapies against neovascular ocular diseases.
Collapse
Affiliation(s)
- Holly C. Cappelli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Brianna D. Guarino
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Anantha K. Kanugula
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Ravi K. Adapala
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Vidushani Perera
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Matthew A. Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- Rebbeca D. Considine Research Institute, Akron Children’s Hospital, Akron, OH 44302
| | | | - Charles K. Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| |
Collapse
|
123
|
Ji G, Li Q, Shen Y, Gan J, Xu L, Wang Y, Luo H, Yang Y, Dong E, Zhang G, Liu B, Yue X, Zhang W, Yang H. Eradication of large established tumors by drug-loaded bacterial particles via a neutrophil-mediated mechanism. J Control Release 2021; 334:52-63. [PMID: 33878368 DOI: 10.1016/j.jconrel.2021.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/13/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
The treatment of large established tumors remains a significant challenge and is generally hampered by poor drug penetration and intrinsic drug resistance of tumor cells in the central tumor region. In the present study, we developed bacterial particles (BactPs) to deliver chemotherapeutics into the tumor mass by hijacking neutrophils as natural cell-based carriers. BactPs loaded with doxorubicin, 5-fluorosuracil, or paclitaxel induced significantly greater tumor regression than unconjugated drugs. This effect was mediated by the ability of BactPs to incorporate chemotherapeutics and serve as vascular disrupting agents that trigger innate host responses and recruit phagocytic neutrophils. Vascular disruption resulted in extensive cell death in the central areas of the tumor mass. Recruited neutrophils acted as natural cellular carriers to deliver engulfed BactPs, which ensured drug delivery into the tumor mass and cytotoxic effects in areas that are normally inaccessible to traditional chemotherapy. Thus, BactPs eradicate large established tumors by functioning as vascular disrupters and natural drug carriers for neutrophil-mediated chemotherapy.
Collapse
Affiliation(s)
- Gaili Ji
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qiqi Li
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yuge Shen
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Jia Gan
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Lin Xu
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, West China Medical School/West China Hospital, Sichuan University, Chengdu, China
| | - Hui Luo
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - E Dong
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Guimin Zhang
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Binrui Liu
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiaozhu Yue
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Wei Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China.
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China; Experimental and Research Animal Institute, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
124
|
Khan MH, Walsh JJ, Mihailović JM, Mishra SK, Coman D, Hyder F. Imaging the transmembrane and transendothelial sodium gradients in gliomas. Sci Rep 2021; 11:6710. [PMID: 33758290 PMCID: PMC7987982 DOI: 10.1038/s41598-021-85925-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/08/2021] [Indexed: 11/29/2022] Open
Abstract
Under normal conditions, high sodium (Na+) in extracellular (Na+e) and blood (Na+b) compartments and low Na+ in intracellular milieu (Na+i) produce strong transmembrane (ΔNa+mem) and weak transendothelial (ΔNa+end) gradients respectively, and these manifest the cell membrane potential (Vm) as well as blood–brain barrier (BBB) integrity. We developed a sodium (23Na) magnetic resonance spectroscopic imaging (MRSI) method using an intravenously-administered paramagnetic polyanionic agent to measure ΔNa+mem and ΔNa+end. In vitro 23Na-MRSI established that the 23Na signal is intensely shifted by the agent compared to other biological factors (e.g., pH and temperature). In vivo 23Na-MRSI showed Na+i remained unshifted and Na+b was more shifted than Na+e, and these together revealed weakened ΔNa+mem and enhanced ΔNa+end in rat gliomas (vs. normal tissue). Compared to normal tissue, RG2 and U87 tumors maintained weakened ΔNa+mem (i.e., depolarized Vm) implying an aggressive state for proliferation, whereas RG2 tumors displayed elevated ∆Na+end suggesting altered BBB integrity. We anticipate that 23Na-MRSI will allow biomedical explorations of perturbed Na+ homeostasis in vivo.
Collapse
Affiliation(s)
- Muhammad H Khan
- Department of Biomedical Engineering, Yale University, N143 TAC (MRRC), 300 Cedar Street, New Haven, CT, 06520, USA.
| | - John J Walsh
- Department of Biomedical Engineering, Yale University, N143 TAC (MRRC), 300 Cedar Street, New Haven, CT, 06520, USA
| | - Jelena M Mihailović
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
| | - Sandeep K Mishra
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
| | - Fahmeed Hyder
- Department of Biomedical Engineering, Yale University, N143 TAC (MRRC), 300 Cedar Street, New Haven, CT, 06520, USA. .,Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
125
|
O'Brien BJ, Singer HA, Adam AP, Ginnan RG. CaMKIIδ is upregulated by pro-inflammatory cytokine IL-6 in a JAK/STAT3-dependent manner to promote angiogenesis. FASEB J 2021; 35:e21437. [PMID: 33749880 DOI: 10.1096/fj.202002755r] [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: 12/22/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 12/17/2022]
Abstract
Ca2+ /calmodulin-dependent protein kinase II (CaMKII) is a ubiquitous serine threonine kinase with established roles in physiological and pathophysiological vascular remodeling. Based on our previous study demonstrating that CaMKIIδ promotes thrombin-induced endothelial permeability and recent reports that CaMKII may contribute to inflammatory remodeling in the heart, we investigated CaMKIIδ-dependent regulation of endothelial function downstream of an interleukin-6 (IL-6)/JAK/STAT3 signaling axis. Upon treatment with IL-6 and its soluble receptor (sIL-6r), CaMKIIδ expression is significantly induced in HUVEC. Using pharmacological inhibitors of JAK and siRNA targeting STAT3, we demonstrated that activation of STAT3 is sufficient to induce CaMKIIδ expression. Under these conditions, rather than promoting IL-6-induced permeability, we found that CaMKIIδ promotes endothelial cell migration as measured by live cell imaging of scratch wound closure and single-cell motility analysis. In a similar manner, endothelial cell proliferation was attenuated upon knockdown of CaMKIIδ as determined by growth curves, cell cycle analysis, and capacitance of cell-covered electrodes as measured by ECIS. Using inducible endothelial-specific STAT3 knockout mice, we demonstrate that STAT3 signaling promotes developmental angiogenesis in the neonatal mouse retina assessed at postnatal day 6. CaMKIIδ expression in retinal endothelium was attenuated in these animals as measured by qPCR. STAT3's effects on angiogenesis were phenocopied by the endothelial-specific knockout of CaMKIIδ, with significantly reduced vascular outgrowth and number of junctions in the developing P6 retina. For the first time, we demonstrate that transcriptional regulation of CaMKIIδ by STAT3 promotes endothelial motility, proliferation, and in vivo angiogenesis.
Collapse
Affiliation(s)
- Brendan J O'Brien
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roman G Ginnan
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
126
|
Zhong L, Pan Y, Shen J. FBXW7 inhibits invasion, migration and angiogenesis in ovarian cancer cells by suppressing VEGF expression through inactivation of β-catenin signaling. Exp Ther Med 2021; 21:514. [PMID: 33791023 PMCID: PMC8005732 DOI: 10.3892/etm.2021.9945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
F-box and WD repeat domain containing 7 (FBXW7) is a tumor suppressor gene frequently inactivated in several human malignancies. The present study aimed to investigate the role of FBXW7 in the invasion, migration and angiogenesis of ovarian cancer (OC) cells, and to identify its potential molecular mechanisms. First, the expression levels of FBXW7 and vascular endothelial growth factor (VEGF) were detected in several human OC cell lines using western blotting. Subsequently, FBXW7 was overexpressed to determine VEGF expression in SKOV3 cells. Transwell, wound healing and tube formation assays were performed following transfection with FBXW7 and VEGF overexpression plasmids to assess invasion, migration and angiogenesis in SKOV3 cells, respectively. Western blot analysis was performed to detect the expression levels of epithelial-to-mesenchymal transition and angiogenesis-associated proteins. In addition, the expression levels of β-catenin and c-Myc were assessed, and lithium chloride (LiCl), an agonist of β-catenin signaling, was used to elucidate the molecular mechanisms by which FBXW7 mediates its antitumor activity in OC. The results demonstrated that FBXW7 expression was markedly downregulated, whilst VEGF expression was markedly upregulated in OC cell lines compared with that in normal ovarian cells. Overexpression of FBXW7 significantly decreased VEGF expression in SKOV3 cells. Notably, overexpression of VEGF reversed the inhibitory effects of FBXW7 overexpression on the invasion, migration and angiogenesis of OC cells, accompanied by upregulated expression levels of N-cadherin, slug, CD31, VEGF receptor 1 (VEGFR1) and VEGFR2, and downregulated expression levels of E-cadherin. Furthermore, overexpression of FBXW7 markedly suppressed β-catenin and c-Myc expression, whereas the decreased expression levels of VEGF, VEGFR1 and VEGFR2 following overexpression of FBXW7 were increased after treatment of SKOV3 cells with LiCl. Overall, the results of the present study suggested that FBXW7 inhibited invasion, migration and angiogenesis of OC cells by suppressing VEGF expression through inactivation of β-catenin signaling. Thus, FBXW7 may be used as a novel therapeutic target for the treatment of OC.
Collapse
Affiliation(s)
- Liping Zhong
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Yuefen Pan
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Junjun Shen
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
127
|
Abstract
BACKGROUND Clubfoot deformity (pes equinovarus) is one of the most common birth defects, and its etiology is still unknown. Initial clubfoot treatment is based on the Ponseti method throughout most of the world. Despite the effectiveness of this therapy, clubfoot may relapse. Recent studies confirm the theory of active fibrotic remodeling processes in the extracellular matrix of the affected tissue. The aim of this study was to clarify whether relapses in clubfoot therapy are associated with altered angiogenesis and to suggest possible regulatory pathways of this pathologic process. METHODS We compared microvessel density, arteriole density, and concentration of angioproliferative-related proteins found between tissues in the contracted, that is, the medial side (M-side), and noncontracted, that is, the lateral side (L-side) of the relapsed clubfeet. Tissue samples from 10 patients were analyzed. Histopathologic analysis consisted of immunohistochemistry and image analysis. Real-time polymerase chain reaction was used to study mRNA expression. RESULTS An increase in microvessel and arteriole density was noted in contracted, relapsed clubfoot tissue. This was accompanied by a significant increase in the levels of the vascular endothelial growth factor, vascular endothelial growth factor receptor 2, β catenin and active β catenin. Vascular endothelial growth factor, vascular endothelial growth factor receptor 2, and CD31 overexpression was also seen with mRNA analysis. CONCLUSIONS Increased microvessel and arteriole density in the contracted side of the relapsed clubfoot was noted. These processes are mediated by specific proangiogenic proteins that are overexpressed in the contracted tissue. These findings contribute to the etiology and the development of relapses in the treatment of clubfoot. LEVEL OF EVIDENCE Level II-analytical and prospective.
Collapse
|
128
|
Chen X, Gao Y, Zhang G, Li B, Ma T, Ma Y, Wang X. Bevacizumab Plays a double-edged role in Neoadjuvant Therapy for Non-metastatic Breast Cancer: A Systemic Review and Meta-Analysis. J Cancer 2021; 12:2643-2653. [PMID: 33854624 PMCID: PMC8040714 DOI: 10.7150/jca.53303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/04/2021] [Indexed: 12/04/2022] Open
Abstract
The anti-angiogenic drug Bevacizumab (Bev) is engaged in neoadjuvant therapy for non-metastatic breast cancer (NMBC). However, whether neoadjuvant Bev providing a greater benefit to patients is debatable. Our study aimed to review Bev's role in Neoadjuvant therapy (NAT) in NMBC and identify predictive markers associated with its efficacy by systemic review and meta-analysis. Eligible trials were retrieved from the Pubmed, Embase, and Cochrane Library, and random or fixed effects models were applied to synthesize data. Power of pCR to predict DFS or OS was evaluated by nonlinear mixed effect model. In NMBC, Bev significantly improved the rate of patients achieving pCR, but this benefit discontinued in DFS or OS. Biomarkers such as PAM50 intrinsic subtype, VEGF overexpression, regulation of VEGF signaling pathway, hypoxia-related genes, BRCA1/2 mutation, P53 mutation and immune phenotype can be used to predict Bev-inducing pCR and/or DFS/OS. Unfortunately, although patients with pCR survived longer than those without pCR when ignoring the use of Bev, but patients achieving pCR with Bev might survive shorter than those achieving pCR without Bev. Subgroup analyses found Bev prolonged patients' OS when given pre- and post-surgery. Lastly, adding Bev increased adverse effects. Overall, Bev offered limited effect for patients with NMBC in an unscreened population. However, in biomarkers - identified subgroup, Bev could be promising to ameliorate the prognosis of specific patients with NMBC.
Collapse
Affiliation(s)
- XinJie Chen
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - Yu Gao
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - GanLin Zhang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - BingXue Li
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - TingTing Ma
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - YunFei Ma
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - XiaoMin Wang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| |
Collapse
|
129
|
Shah AM, Jain K, Desai RS, Bansal S, Shirsat P, Prasad P, Bodhankar K. The Role of Increased Connective Tissue Growth Factor in the Pathogenesis of Oral Submucous Fibrosis and its Malignant Transformation-An Immunohistochemical Study. Head Neck Pathol 2021; 15:817-830. [PMID: 33544386 PMCID: PMC8384978 DOI: 10.1007/s12105-020-01270-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Connective tissue growth factor (CTGF), a matricellular protein of the CCN family of extracellular matrix-associated heparin-binding proteins, is highly expressed in various organ fibrosis and several malignant tumors. Although a few studies have been conducted using CTGF in oral submucous fibrosis (OSF) and oral squamous cell carcinoma, no study has demonstrated its relation with various stages of OSF and its malignant transformation. The present study investigated the possible role of CTGF in the pathogenesis of OSF and its malignant transformation by using immunohistochemistry. Ten formalin-fixed paraffin-embedded tissue blocks, each of Stage 1 OSF, Stage 2 OSF, Stage 3 OSF, Stage 4 OSF, well- differentiated squamous cell carcinoma (WDSCC) with OSF and WDSCC without OSF were stained for CTGF by immunohistochemistry. Ten cases of healthy buccal mucosa (NOM) were included as controls. The present study demonstrated a statistically significant expression of CTGF in the epithelium and connective tissue of OSF and WDSCC with and without OSF cases against its complete absence in NOM. We observed an upregulation of CTGF expression from NOM to various stages of OSF to WDSCC with or without OSF. A gradual upregulation of the CTGF expression in various stages of OSF to WDSCC (with and without OSF) against its complete absence in NOM suggests that CTGF plays an important role in the pathogenesis of OSF and its malignant transformation.
Collapse
Affiliation(s)
| | - Kejal Jain
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| | - Rajiv S. Desai
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| | - Shivani Bansal
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| | - Pankaj Shirsat
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| | - Pooja Prasad
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| | - Kshitija Bodhankar
- Department of Oral Pathology, Nair Hospital Dental College, Mumbai, 400008 India
| |
Collapse
|
130
|
Pawar NV, Singh PD, Prabhu PS, Rana JR. Carcinogen-Induced Model of Proangiogenesis in Zebrafish Embryo-Larvae. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:447-453. [PMID: 33179787 DOI: 10.1002/etc.4928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/30/2020] [Accepted: 11/09/2020] [Indexed: 06/11/2023]
Abstract
Tumor angiogenesis is the main target in cancer drug development. Discovery of antiangiogenic agents targeting different mechanisms of action is the major area of research to control tumor growth and metastasis. Zebrafish (in the embryo-larvae stage) acts as an essential preclinical efficacy-toxicity model for antiangiogenic drug discovery. We aimed to develop a carcinogen-induced model of proangiogenesis in zebrafish embryo-larvae using the carcinogens lindane and benzo[a]pyrene. Zebrafish were randomly selected for mating. Postspawning, healthy embryos were staged, dispensed in reverse-osmosis water in a 12-well plate, and incubated at 28.5 °C, wherein 24 h postfertilization they were exposed to sublethal concentrations of the carcinogens. Three days postexposure, embryos were stained with alkaline phosphatase, and the angiogenic basket was imaged using a bright-field microscope. The number of subintestinal vessels, their length from somite to the basket, and other proangiogenic parameters were measured and analyzed. The effective concentrations causing a 30% increase in subintestinal vessels for benzo[a]pyrene and lindane were 2.69 and 2.24 µM, respectively, thus proving their proangiogenic potency. The carcinogen-induced model of proangiogenesis in zebrafish embryo-larvae can be used as an effective high-throughput screening tool to assess the proangiogenic potential of carcinogenic compounds and to screen antiangiogenic drugs for better therapeutic intervention. Environ Toxicol Chem 2021;40:447-453.© 2020 SETAC.
Collapse
Affiliation(s)
- Nilambari V Pawar
- Section of Ecotoxicology, Department of Chemistry, Jai Research Foundation, Valvada, Gujarat, India
| | - Pritee D Singh
- Section of Ecotoxicology, Department of Chemistry, Jai Research Foundation, Valvada, Gujarat, India
| | - Padmaja S Prabhu
- Section of Ecotoxicology, Department of Chemistry, Jai Research Foundation, Valvada, Gujarat, India
| | - Jigarkumar R Rana
- Section of Ecotoxicology, Department of Chemistry, Jai Research Foundation, Valvada, Gujarat, India
| |
Collapse
|
131
|
Ridwan SM, Hainfeld JF, Ross V, Stanishevskiy Y, Smilowitz HM. Novel Iodine nanoparticles target vascular mimicry in intracerebral triple negative human MDA-MB-231 breast tumors. Sci Rep 2021; 11:1203. [PMID: 33441981 PMCID: PMC7806637 DOI: 10.1038/s41598-020-80862-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/23/2020] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC), ~ 10-20% of diagnosed breast cancers, metastasizes to brain, lungs, liver. Iodine nanoparticle (INP) radioenhancers specifically localize to human TNBC MDA-MB-231 tumors growing in mouse brains after iv injection, significantly extending survival of mice after radiation therapy (RT). A prominent rim of INP contrast (MicroCT) previously seen in subcutaneous tumors but not intracerebral gliomas, provide calculated X-ray dose-enhancements up to > eightfold. Here, MDA-MB-231-cells, INPs, CD31 were examined by fluorescence confocal microscopy. Most INP staining co-localized with CD31 in the tumor center and periphery. Greatest INP/CD31 staining was in the tumor periphery, the region of increased MicroCT contrast. Tumor cells are seen to line irregularly-shaped spaces (ISS) with INP, CD31 staining very close to or on the tumor cell surface and PAS stain on their boundary and may represent a unique form of CD31-expressing vascular mimicry in intracerebral 231-tumors. INP/CD31 co-staining is also seen around ISS formed around tumor cells migrating on CD31+ blood-vessels. The significant radiation dose enhancement to the prolific collagen I containing, INP-binding ISS found throughout the tumor but concentrated in the tumor rim, may contribute significantly to the life extensions observed after INP-RT; VM could represent a new drug/NP, particularly INP, tumor-homing target.
Collapse
Affiliation(s)
- Sharif M Ridwan
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | | | - Vanessa Ross
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | | | - Henry M Smilowitz
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
132
|
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: 345] [Impact Index Per Article: 86.3] [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
|
133
|
Antileishmanial Activity of Lignans, Neolignans, and Other Plant Phenols. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2021; 115:115-176. [PMID: 33797642 DOI: 10.1007/978-3-030-64853-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Secondary metabolites (SM) from organisms have served medicinal chemists over the past two centuries as an almost inexhaustible pool of new drugs, drug-like skeletons, and chemical probes that have been used in the "hunt" for new biologically active molecules with a "beneficial effect on human mind and body." Several secondary metabolites, or their derivatives, have been found to be the answer in the quest to search for new approaches to treat or even eradicate many types of diseases that oppress humanity. A special place among SM is occupied by lignans and neolignans. These phenolic compounds are generated biosynthetically via radical coupling of two phenylpropanoid monomers, and are known for their multitarget activity and low toxicity. The disadvantage of the relatively low specificity of phenylpropanoid-based SM turns into an advantage when structural modifications of these skeletons are made. Indeed, phenylpropanoid-based SM previously have proven to offer great potential as a starting point in drug development. Compounds such as Warfarin® (a coumarin-based anticoagulant) as well as etoposide and teniposide (podophyllotoxin-based anticancer drugs) are just a few examples. At the beginning of the third decade of the twenty-first century, the call for the treatment of more than a dozen rare or previously "neglected" diseases remains for various reasons unanswered. Leishmaniasis, a neglected disease that desperately needs new ways of treatment, is just one of these. This disease is caused by more than 20 leishmanial parasites that are pathogenic to humans and are spread by as many as 800 sandfly species across subtropical areas of the world. With continuing climate changes, the presence of Leishmania parasites and therefore leishmaniasis, the disease caused by these parasites, is spreading from previous locations to new areas. Thus, leishmaniasis is affecting each year a larger proportion of the world's population. The choice of appropriate leishmaniasis treatment depends on the severity of the disease and its form of manifestation. The success of current drug therapy is often limited, due in most cases to requiring long hospitalization periods (weeks to months) and the toxicity (side effects) of administered drugs, in addition to the increasing resistance of the parasites to treatment. It is thus important to develop new drugs and treatments that are less toxic, can overcome drug resistance, and require shorter periods of treatment. These aspects are especially important for the populations of developing countries. It was reported that several phenylpropanoid-based secondary metabolites manifest interesting antileishmanial activities and are used by various indigenous people to treat leishmaniasis. In this chapter, the authors shed some light on the various biological activities of phenylpropanoid natural products, with the main focus being on their possible applications in the context of antileishmanial treatment.
Collapse
|
134
|
Augimeri G, Gelsomino L, Plastina P, Giordano C, Barone I, Catalano S, Andò S, Bonofiglio D. Natural and Synthetic PPARγ Ligands in Tumor Microenvironment: A New Potential Strategy against Breast Cancer. Int J Mol Sci 2020; 21:E9721. [PMID: 33352766 PMCID: PMC7767156 DOI: 10.3390/ijms21249721] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple lines of evidence indicate that activation of the peroxisome proliferator-activated receptor γ (PPARγ) by natural or synthetic ligands exerts tumor suppressive effects in different types of cancer, including breast carcinoma. Over the past decades a new picture of breast cancer as a complex disease consisting of neoplastic epithelial cells and surrounding stroma named the tumor microenvironment (TME) has emerged. Indeed, TME is now recognized as a pivotal element for breast cancer development and progression. Novel strategies targeting both epithelial and stromal components are under development or undergoing clinical trials. In this context, the aim of the present review is to summarize PPARγ activity in breast TME focusing on the role of this receptor on both epithelial/stromal cells and extracellular matrix components of the breast cancer microenvironment. The information provided from the in vitro and in vivo research indicates PPARγ ligands as potential agents with regards to the battle against breast cancer.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
| | - Pierluigi Plastina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| |
Collapse
|
135
|
Liu K, Zhang X, Xie L, Deng M, Chen H, Song J, Long J, Li X, Luo J. Lupeol and its derivatives as anticancer and anti-inflammatory agents: Molecular mechanisms and therapeutic efficacy. Pharmacol Res 2020; 164:105373. [PMID: 33316380 DOI: 10.1016/j.phrs.2020.105373] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
Lupeol is a natural triterpenoid that widely exists in edible fruits and vegetables, and medicinal plants. In the last decade, a plethora of studies on the pharmacological activities of lupeol have been conducted and have demonstrated that lupeol possesses an extensive range of pharmacological activities such as anticancer, antioxidant, anti-inflammatory, and antimicrobial activities. Pharmacokinetic studies have indicated that absorption of lupeol by animals was rapid despite its nonpolar characteristics, and lupeol belongs to class II BCS (biopharmaceutics classification system) compounds. Moreover, the bioactivities of some isolated or synthesized lupeol derivatives have been investigated, and these results showed that, with modification to C-3 or C-19, some derivatives exhibit stronger activities, e.g., antiprotozoal or anticancer activity. This review aims to summarize the advances in pharmacological and pharmacokinetic studies of lupeol in the last decade with an emphasis on its anticancer and anti-inflammatory activities, as well as the research progress of lupeol derivatives thus far, to provide researchers with the latest information, point out the limitations of relevant research at the current stage and the aspects that should be strengthened in future research.
Collapse
Affiliation(s)
- Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xumin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Mao Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Huijuan Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiawen Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiaying Long
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Jia Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
136
|
Zommiti M, Feuilloley MGJ, Connil N. Update of Probiotics in Human World: A Nonstop Source of Benefactions till the End of Time. Microorganisms 2020; 8:E1907. [PMID: 33266303 PMCID: PMC7760123 DOI: 10.3390/microorganisms8121907] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Lactic acid bacteria (LAB) are known for their biotechnological potential. Moreover, LAB are distinguished by amazing criteria: Adjusting the intestinal environment, inhibiting pathogenic microbes in the gastrointestinal tract, ability to reduce pathogen adhesion activity, improving the balance of the microbiota inside the intestine, capabilities of regulating intestinal mucosal immunity, and maintaining intestinal barrier function. The escalating number of research and studies about beneficial microorganisms and their impact on promoting health has attracted a big interest in the last decades. Since antiquity, various based fermented products of different kinds have been utilized as potential probiotic products. Nevertheless, the current upsurge in consumers' interest in bioalternatives has opened new horizons for the probiotic field in terms of research and development. The present review aims at shedding light on the world of probiotics, a continuous story of astonishing success in various fields, in particular, the biomedical sector and pharmaceutical industry, as well as to display the importance of probiotics and their therapeutic potential in purpose to compete for sturdy pathogens and to struggle against diseases and acute infections. Shadows and future trends of probiotics use are also discussed.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| |
Collapse
|
137
|
Aventurado CA, Billones JB, Vasquez RD, Castillo AL. In Ovo and In Silico Evaluation of the Anti-Angiogenic Potential of Syringin. Drug Des Devel Ther 2020; 14:5189-5204. [PMID: 33268982 PMCID: PMC7701684 DOI: 10.2147/dddt.s271952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/08/2020] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Cancer is considered as one of the deadliest human diseases today. Angiogenesis, the propagation of new blood vessels from pre-existing vasculature, is a critical step in the progression of cancer as it is essential in the growth and metastasis of tumors. Hence, suppression of angiogenesis is a promising approach in cancer therapy. Syringin, a phenylpropanoid glycoside with a molecular formula of C17H24O9, has been found to exhibit chemopreventive effects. However, its anti-angiogenic activity and the underlying mechanism of action are still unknown. METHODS In this work, in ovo chorioallantoic membrane (CAM) assay has been conducted to evaluate the effect of syringin on neovascularization. Additionally, reverse molecular docking studies have been performed in order to identify the probable enzyme targets in the angiogenesis pathway. RESULTS Treatment with syringin showed significant dose-dependent inhibition of blood vessel length and junctions in the CAM of duck eggs; the anti-angiogenic activity of syringin at 100 µM and 200 µM is comparable with 200 µM of the positive control celecoxib. The results of reverse docking studies indicate that syringin binds the strongest to dihydrofolate reductase (DHFR) and, to some extent, with transforming growth factor-beta receptor type 1 (TGF-βR1), vascular endothelial growth factor receptor 2 (VEGFR2), and matrix metalloproteinase-2 (MMP-2). Furthermore, ADMET models revealed that syringin potentially possesses excellent pharmacokinetic and toxicity profiles. CONCLUSION This study demonstrates the potential of syringin as an anti-angiogenic agent and elicits further investigations to establish its application in cancer suppression.
Collapse
Affiliation(s)
| | - Junie B Billones
- Department of Physical Sciences and Mathematics, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| | - Ross D Vasquez
- The Graduate School, University of Santo Tomas, Manila1015, Philippines
- Faculty of Pharmacy, University of Santo Tomas, Manila1015, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila1015, Philippines
| | - Agnes L Castillo
- The Graduate School, University of Santo Tomas, Manila1015, Philippines
- Faculty of Pharmacy, University of Santo Tomas, Manila1015, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila1015, Philippines
| |
Collapse
|
138
|
Arjmand F, Khursheed S, Roisnel T, Siddique HR. Copper (II)-based halogen-substituted chromone antitumor drug entities: Studying biomolecular interactions with ct-DNA mediated by sigma hole formation and cytotoxicity activity. Bioorg Chem 2020; 104:104327. [PMID: 33142405 DOI: 10.1016/j.bioorg.2020.104327] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 12/26/2022]
Abstract
Copper-based antitumor drug entities 1-3 derived from substituted (F-, Br-, -CH3) 3-formylchromone pharmacophore were synthesized and thoroughly characterized by spectroscopic and single X-ray crystallographic studies. These complexes show structural novelty due to presence of the X-bonds in chromone scaffold which could facilitate higher propensity for nucleic acids via sigma σ-hole interactions. Therefore, structure-activity relationship of 1-3 was studied by performing ct-DNA binding, pBR322 cleavage and cytotoxicity activity to validate their potential to act as chemotherapeutic drug entities. The binding studies of 1-3 with ct- DNA were carried out employing many biophysical techniques and the corroborative results of these experiments showed intercalation mode of binding and the order of binding was found to be 2 > 1 > 3. The structure of drug entities could facilitated strong halogen bonding interaction (in case of 1 &2) and stability of X bond was rationalized by sigma hole region of positive electrostatic potential on the surface of C-X covalent bond, as determined by gas phase B3LYP computational DFT studies. Interestingly, 2 exhibited most avid binding affinity due to presence of Br- electron withdrawing and polarizable group. Further, cleavage studies of 1-3 with pBR322 plasmid DNA were performed which demonstrated significant cleavage activity, the supercoiled form (Form I) of plasmid DNA was converted to nicked form (Form II) with the appearance of linearized form (Form III) in between two, implicating lethal double strand breaks of DNA. 2 showed predominantly higher cleavage activity following the similar trend as observed for binding studies. The cytotoxicity of the complexes 1-3 was evaluated by MTT assay against the human liver carcinoma (Huh-7) and prostate cancer (DU-145) cell lines; complex 2 exhibited specific and selective cytotoxicity for the DU-145 cancer cell line with LC50 value of 1.6 μM.
Collapse
Affiliation(s)
- Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University, Aligarh, India.
| | - Salman Khursheed
- Department of Chemistry, Aligarh Muslim University, Aligarh, India
| | - Thierry Roisnel
- Institut des Sciences Chimiques de Rennes, UMR 6226, Universit́e de Rennes 1, Campus de Beaulieu Batiment 10B, Bureau, 15335042 Rennes, France
| | - Hifzur R Siddique
- Cytogenetics and Molecular Toxicology Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| |
Collapse
|
139
|
Abdelgawad ME, Darwish H, Nabawy MM, El-mezayen H. Development of novel score based on Angiogenic panel for accurate diagnosis of hepatocellular carcinoma among hepatitis C virus high-risk patients. INFECTION GENETICS AND EVOLUTION 2020; 85:104572. [DOI: 10.1016/j.meegid.2020.104572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
|
140
|
Feng H, Liu K, Shen X, Liang J, Wang C, Qiu W, Cheng X, Zhao R. Targeting tumor cell-derived CCL2 as a strategy to overcome Bevacizumab resistance in ETV5 + colorectal cancer. Cell Death Dis 2020; 11:916. [PMID: 33099574 PMCID: PMC7585575 DOI: 10.1038/s41419-020-03111-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
In our previous study, ETV5 mediated-angiogenesis was demonstrated to be dependent upon the PDGF-BB/PDGFR-β/Src/STAT3/VEGFA pathway in colorectal cancer (CRC). However, the ability of ETV5 to affect the efficacy of anti-angiogenic therapy in CRC requires further investigation. Gene set enrichment analysis (GSEA) and a series of experiments were performed to identify the critical candidate gene involved in Bevacizumab resistance. Furthermore, the ability of treatment targeting the candidate gene to enhance Bevacizumab sensitivity in vitro and in vivo was investigated. Our results revealed that ETV5 directly bound to the VEGFA promoter to promote translation of VEGFA. However, according to in vitro and in vivo experiments, ETV5 unexpectedly accelerated antiVEGF therapy (Bevacizumab) resistance. GSEA and additional assays confirmed that ETV5 could promote angiogenesis by inducing the secretion of another tumor angiogenesis factor (CCL2) in CRC cells to facilitate Bevacizumab resistance. Mechanistically, ETV5 upregulated CCL2 by activating STAT3 to facilitate binding with the CCL2 promoter. ETV5 induced-VEGFA translation and CCL2 secretion were mutually independent mechanisms, that induced angiogenesis by activating the PI3K/AKT and p38/MAPK signaling pathways in human umbilical vein endothelial cells (HUVECs). In CRC tissues, ETV5 protein levels were positively associated with CD31, CCL2, and VEGFA protein expression. CRC patients possessing high expression of ETV5/VEGFA or ETV5/CCL2 exhibited a poorer prognosis compared to that of other patients. Combined antiCCL2 and antiVEGFA (Bevacizumab) treatment could inhibit tumor angiogenesis and growth more effectively than single treatments in CRCs with high expression of ETV5 (ETV5+ CRCs). In conclusion, our results not only revealed ETV5 as a novel biomarker for anti-angiogenic therapy, but also indicated a potential combined therapy strategy that involved in targeting of both CCL2 and VEGFA in ETV5+ CRC.
Collapse
Affiliation(s)
- Haoran Feng
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China
| | - Kun Liu
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, 201800, Shanghai, China
| | - Xiaonan Shen
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Juyong Liang
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China
| | - Changgang Wang
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, 201800, Shanghai, China
| | - Weihua Qiu
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Department of General Surgery, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, 201800, Shanghai, China.
| | - Xi Cheng
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Department of General Surgery, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, 201800, Shanghai, China.
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China. .,Department of General Surgery, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, 201800, Shanghai, China.
| |
Collapse
|
141
|
Mortensen OE, Nerup N, Thorsteinsson M, Svendsen MBS, Shiwaku H, Achiam MP. Fluorescence guided intraluminal endoscopy in the gastrointestinal tract: A systematic review. World J Gastrointest Endosc 2020; 12:388-400. [PMID: 33133375 PMCID: PMC7579525 DOI: 10.4253/wjge.v12.i10.388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/29/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Conventional endoscopy is based on full spectrum white light. However, different studies have investigated the use of fluorescence based endoscopy systems where the white light has been supplemented by infrared light and the use of relevant fluorophores. Fluorescence endoscopy utilizes the fluorescence emitted from a fluorophore, visualizing what is not visible to the naked eye.
AIM To explore the feasibility of fluorescence endoscopy and evaluate its use in diagnosing and evaluating gastrointestinal disease.
METHODS We followed the PRISMA guidelines for this systematic review. The research covered five databases; PubMed, Scopus, Web of Science, Embase, and the Cochrane Collection, including only studies in English and Scandinavian languages. Authors screened title and abstract for inclusion, subsequently full-text for inclusion according to eligibility criteria listed in the protocol. The risk of bias was assessed for all studies according to the Newcastle-Ottawa Scale. The authors extracted the data and reported the results in both text and tables.
RESULTS We included seven studies in the systematic review after screening a total of 2769 papers. The most prominent fluorophore was indocyanine green (n = 6), and whereas one study (n = 1) used Bevacizumab 800-CW. Three studies investigated fluorescence endoscopy in detecting varices, adenomas in patients with familial adenomatous polyposis and neoplasms in the gastrointestinal tract. Four studies evaluated the usefulness of fluorescence endoscopy in assessing tumor invasion. Three of the four studies reported an exceptional diagnostic accuracy (93%, 89% and 88%) in assessing tumor invasion, thus representing better visualization and more correct diagnosis by fluorescence endoscopy compared with the conventional endoscopy. The relationship between the endoscopic findings, tumor invasion, and tumor vascularity was evaluated in two studies showing a significant correlation (dP < 0.05 and bP < 0.01).
CONCLUSION The use of fluorescence endoscopy is a promising method adding diagnostic value in the detection of neoplasia, adenomas, and assessment of tumor invasion within the gastrointestinal tract. More studies are needed to utilize the feasibility of fluorescence endoscopy compared with other endoscopic methods.
Collapse
Affiliation(s)
- Olivia Engholt Mortensen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Nikolaj Nerup
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Morten Thorsteinsson
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | | | - Hironari Shiwaku
- Department of Surgical Gastroenterology, Fukuoka University Faculty of Medicine, Fukuoka 814-0133, Japan
| | - Michael Patrick Achiam
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Copenhagen 2100, Denmark
| |
Collapse
|
142
|
Wang D, Xiao F, Feng Z, Li M, Kong L, Huang L, Wei Y, Li H, Liu F, Zhang H, Zhang W. Sunitinib facilitates metastatic breast cancer spreading by inducing endothelial cell senescence. Breast Cancer Res 2020; 22:103. [PMID: 32993785 PMCID: PMC7526390 DOI: 10.1186/s13058-020-01346-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/21/2020] [Indexed: 02/08/2023] Open
Abstract
Background Sunitinib, a receptor tyrosine kinase (RTK) inhibitor that targets multiple receptors such as vascular endothelial growth factor receptors (VEGFRs), was approved for cancer treatment in 2006. However, it was unsuccessful in treating certain cancers, particularly metastatic breast cancer (MBC), and the mechanism underlying this “sunitinib resistance” remains unclear. Herein, we investigated whether the sunitinib-associated inferior survival benefit in MBC was due to sunitinib-induced endothelial cell (EC) injury or EC senescence. Methods 4T1 murine breast cancer cells were used as the main breast tumor model for it produces a highly metastatic solid tumor that can spontaneously metastasize to the lung, which closely mimics highly metastatic human breast cancer. Senescence-associated β-galactosidase (SA-β-Gal, immunohistochemistry [IHC]-staining), P16, P53, and P57 (immunoblotting) were used as markers of cell senescence. A protein array containing 25 senescence-associated chemokines and the transwell chemotaxis assay were used to examine whether sunitinib increases inflammatory chemokine secretion which attracts tumor cells via chemokinesis. Flow cytometry and IHC were used to detect whether the sunitinib-induced senescent ECs recruit cancer-associated inflammatory myeloid cells. Finally, the spontaneous metastatic model was used to monitor whether sunitinib causes the formation of “pre-metastatic niche” which promotes MBC to metastasize to the lungs. Results We demonstrated that sunitinib induced a senescence-like endothelial cell (EC) phenotype. Inflammatory chemokine secretion and VCAM1 expression were significantly increased in senescent ECs, resulting in tumor cell (TC) chemotaxis and TC/EC interactions. Meanwhile, EC senescence caused loosening of EC junctions, facilitating TC transmigration through the endothelial barrier. Sunitinib-induced senescent ECs also recruited cancer-associated myeloid cells to form a “pre-metastatic niche”-like microenvironment. Alterations at the molecular level and in the tissue environment ultimately led to an increase in distant metastasis. Conclusion Although sunitinib was designed to target the EC directly, the increase in tumor metastasis may ironically be due to sunitinib “correctly” playing its role. Our findings suggest that we should carefully weigh the pros and cons before using sunitinib and other antiangiogenic drugs that directly target the ECs.
Collapse
Affiliation(s)
- Denian Wang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhongxue Feng
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Min Li
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lingmiao Kong
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Luping Huang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong'gang Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hongyu Li
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Fei Liu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Haili Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wei Zhang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
143
|
In vivo longitudinal and multimodal imaging of hypoxia-inducible factor 1α and angiogenesis in breast cancer. Chin Med J (Engl) 2020; 133:205-211. [PMID: 31904728 PMCID: PMC7028170 DOI: 10.1097/cm9.0000000000000616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background Angiogenesis and hypoxia-inducible factor 1α (HIF-1α) play major roles in solid tumors. This study aimed to establish a longitudinal and multimodal imaging model for in vivo evaluation of HIF1α and angiogenesis in breast cancer. Methods By transfection of a 5 hypoxia-responsive element (HRE)/green fluorescent protein (GFP) plasmid, the cell line Ca761-hre-gfp was established, which emitted green fluorescence triggered by HIF-1α under hypoxia. The cells were subjected to CoCl2-simulated hypoxia to confirm the imaging strategy. We grew Ca761-hre-gfp cells in the left rear flanks of twelve 615 mice. Experiments were conducted on days 4, 9, 15, and 19. For in vivo analysis, Ca761-hre-gfp subcutaneous allografted tumors were imaged in vivo using contrast-enhanced ultrasound (CEUS) and fluorescence imaging (FLI) during tumor development. The tumor size, CEUS peak intensity, and FLI photons were measured to evaluate tumor growth, angiogenesis, and HIF-1α activity, respectively. After each experiment, three mice were randomly sacrificed and tumor specimens were collected to examine HIF-1α activity and the microvessel density (MVD). Results In vitro, both green fluorescence and HIF-1α expression were detected in Ca761-hre-gfp cells treated with CoCl2, indicating the suitability of the cells to detect HIF-1α activity. In vivo, HIF-1α activity first increased and then decreased, which was significantly correlated with angiogenic changes (r = 0.803, P = 0.005). These changes were confirmed by immunohistochemical staining of HIF-1α and MVD. Conclusions The findings validated the Ca761-hre-gfp murine allograft model for reliable evaluation of HIF-1α activity and angiogenesis longitudinally using both molecular and pre-clinical non-invasive imaging modalities. The cell line may be useful for studies of anti-HIF pathway therapies.
Collapse
|
144
|
Lamuraglia M, Barrois G, Le Guillou-Buffello D, Santin M, Kerbol A, Comperat E, Coron A, Lucidarme O, Bridal SL. Monitoring Dual VEGF Inhibition in Human Pancreatic Tumor Xenografts With Dynamic Contrast-Enhanced Ultrasound. Technol Cancer Res Treat 2020; 19:1533033819886896. [PMID: 32065066 PMCID: PMC7026814 DOI: 10.1177/1533033819886896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Association of drugs acting against different antiangiogenic mechanisms may increase therapeutic effect and reduce resistance. Noninvasive monitoring of changes in the antiangiogenic response of individual tumors could guide selection and administration of drug combinations. Noninvasive detection of early therapeutic response during dual, vertical targeting of the vascular endothelial growth factor pathway was investigated in an ectopic subcutaneous xenograft model for human pancreatic tumor. METHODS Dynamic contrast-enhanced ultrasound 12 MHz was used to monitor tumor-bearing Naval Medical Research Institute mice beginning 15 days after tumor implantation. Mice received therapy from 15 to 29 days with sorafenib (N = 9), ziv-aflibercept (N = 11), combined antiangiogenic agents (N = 11), and placebo control (N = 14). Sorafenib (BAY 43-9006; Nexavar), a multikinase inhibitor acting on Raf kinase and receptor tyrosine kinases-including vascular endothelial growth factor receptors 2 and 3-was administered daily (60 mg/kg, per os). Ziv-aflibercept (ZALTRAP), a high-affinity ligand trap blocking the activity of vascular endothelial growth factor A, vascular endothelial growth factor B, and placental growth factor was administered twice per week (40 mg/kg, intraperitoneally). RESULTS Functional evaluation with dynamic contrast-enhanced ultrasound indicated stable tumor vascularization for the control group while revealing significant and sustained reduction after 1 day of therapy in the combined group (P = .007). There was no survival benefit or penalty due to drug combination. The functional progression-free survival assessed with dynamic contrast-enhanced ultrasound was significantly higher for the 3 treated groups; whereas, the progression-free survival based on tumor size did not discriminate therapeutic effect. CONCLUSIONS Dynamic contrast-enhanced ultrasound, therefore, presents strong potential to monitor microvascular modifications during antiangiogenic therapy, a key role to monitoring antiangiogenic combining therapy to adapt dose range drug.
Collapse
Affiliation(s)
- Michele Lamuraglia
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Beaujon, Paris, France
| | - Guillaume Barrois
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | | | - Mathieu Santin
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Anne Kerbol
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Beaujon, Paris, France
| | - Eva Comperat
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Alain Coron
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Olivier Lucidarme
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - S Lori Bridal
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| |
Collapse
|
145
|
Ruthenium(II)/(III) DMSO-Based Complexes of 2-Aminophenyl Benzimidazole with In Vitro and In Vivo Anticancer Activity. Molecules 2020; 25:molecules25184284. [PMID: 32962014 PMCID: PMC7570852 DOI: 10.3390/molecules25184284] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
New anticancer ruthenium(II/III) complexes [RuCl2(DMSO)2(Hapbim)] (1) and [RuCl3(DMSO) (Hapbim)] (2) (Hapbim = 2-aminophenyl benzimidazole) have been synthesized and characterized, and their chemotherapeutic potential evaluated. The interaction of the compounds with DNA was studied by both UV-Visible and fluorescence spectroscopies, revealing intercalation of both the Hapbim ligand and the Ru complexes. The in vitro cytotoxicity of the compounds was tested on human breast cancer (MCF7), human colorectal cancer (Caco2), and normal human liver cell lines (THLE-2), with compound (2) the most potent against cancer cells. The cytotoxic effect of (2) is shown to correlate with the ability of the Ru(III) complex to induce apoptosis and to cause cell-cycle arrest in the G2/M phase. Notably, both compounds were inactive in the noncancerous cell line. The anticancer effect of (2) has also been studied in an EAC (Ehrlich Ascites Carcinoma) mouse model. Significantly, the activity of the complex was more pronounced in vivo, with removal of the cancer burden at doses that resulted in only low levels of hepatotoxicity and nephrotoxicity. An apoptosis mechanism was determined by the observation of increased Bax and caspase 3 and decreased Bcl2 expression. Furthermore, (2) decreased oxidative stress and increased the levels of antioxidant enzymes, especially SOD, suggesting the enhancement of normal cell repair. Overall, compound (2) shows great potential as a chemotherapeutic candidate, with promising activity and low levels of side effects.
Collapse
|
146
|
Luengas-Martinez A, Hardman-Smart J, Paus R, Young HS. Vascular endothelial growth factor-A as a promising therapeutic target for the management of psoriasis. Exp Dermatol 2020; 29:687-698. [PMID: 32654325 DOI: 10.1111/exd.14151] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factor-A (VEGF-A), the main angiogenic mediator, plays a critical role in the pathogenesis of several inflammatory immune-mediated diseases, including psoriasis. Even though anti-angiogenic therapies, such as VEGF inhibitors, are licensed for the treatment of various cancers and eye disease, VEGF-targeting interventions are not part of current psoriasis therapy. In this viewpoint essay, we argue that the existing preclinical research evidence on the role of VEGF-A in the pathogenesis of psoriasis as well as clinical observations in patients who have experienced psoriasis remission during oncological anti-VEGF-A therapy strongly suggests to systematically explore angiogenesis targeting also in the management of psoriasis. We also point out that some psoriasis therapies decrease circulating levels of VEGF-A and normalise the psoriasis-associated vascular pathology in the papillary dermis of plaques of psoriasis and that a subset of patients with constitutionally high levels of VEGF-A may benefit most from the anti-angiogenic therapy we advocate here. Given that novel, well-targeted personalised medicine therapies for the development of psoriasis need to be developed, we explore the hypothesis that VEGF-A and signalling through its receptors constitute a promising target for therapeutic intervention in the future management of psoriasis.
Collapse
Affiliation(s)
- Andrea Luengas-Martinez
- Centre for Dermatology Research and Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | | | - Ralf Paus
- Centre for Dermatology Research and Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.,Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Monasterium Laboratory, Muenster, Germany
| | - Helen S Young
- Centre for Dermatology Research and Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
147
|
Mandrycky C, Hadland B, Zheng Y. 3D curvature-instructed endothelial flow response and tissue vascularization. SCIENCE ADVANCES 2020; 6:eabb3629. [PMID: 32938662 PMCID: PMC7494348 DOI: 10.1126/sciadv.abb3629] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/21/2020] [Indexed: 05/08/2023]
Abstract
Vascularization remains a long-standing challenge in engineering complex tissues. Particularly needed is recapitulating 3D vascular features, including continuous geometries with defined diameter, curvature, and torsion. Here, we developed a spiral microvessel model that allows precise control of curvature and torsion and supports homogeneous tissue perfusion at the centimeter scale. Using this system, we showed proof-of-principle modeling of tumor progression and engineered cardiac tissue vascularization. We demonstrated that 3D curvature induced rotation and mixing under laminar flow, leading to unique phenotypic and transcriptional changes in endothelial cells (ECs). Bulk and single-cell RNA-seq identified specific EC gene clusters in spiral microvessels. These mark a proinflammatory phenotype associated with vascular development and remodeling, and a unique cell cluster expressing genes regulating vascular stability and development. Our results shed light on the role of heterogeneous vascular structures in differential development and pathogenesis and provide previously unavailable tools to potentially improve tissue vascularization and regeneration.
Collapse
Affiliation(s)
- Christian Mandrycky
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Center for Cardiovascular Biology, and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
- Center for Cardiovascular Biology, and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
148
|
Potential role of hydrogen sulfide in diabetes-impaired angiogenesis and ischemic tissue repair. Redox Biol 2020; 37:101704. [PMID: 32942144 PMCID: PMC7498944 DOI: 10.1016/j.redox.2020.101704] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is one of the most prevalent metabolic disorders and is estimated to affect 400 million of 4.4% of population worldwide in the next 20 year. In diabetes, risk to develop vascular diseases is two-to four-fold increased. Ischemic tissue injury, such as refractory wounds and critical ischemic limb (CLI) are major ischemic vascular complications in diabetic patients where oxygen supplement is insufficient due to impaired angiogenesis/neovascularization. In spite of intensive studies, the underlying mechanisms of diabetes-impaired ischemic tissue injury remain incompletely understood. Hydrogen sulfide (H2S) has been considered as a third gasotransmitter regulating angiogenesis under physiological and ischemic conditions. Here, the underlying mechanisms of insufficient H2S-impaired angiogenesis and ischemic tissue repair in diabetes are discussed. We will primarily focuses on the signaling pathways of H2S in controlling endothelial function/biology, angiogenesis and ischemic tissue repair in diabetic animal models. We summarized that H2S plays an important role in maintaining endothelial function/biology and angiogenic property in diabetes. We demonstrated that exogenous H2S may be a theraputic agent for endothelial dysfunction and impaired ischemic tissue repair in diabetes.
Collapse
|
149
|
Suzuki T, Tanaka M, Sasaki M, Ichikawa H, Nishie H, Kataoka H. Vascular Shutdown by Photodynamic Therapy Using Talaporfin Sodium. Cancers (Basel) 2020; 12:cancers12092369. [PMID: 32825648 PMCID: PMC7563359 DOI: 10.3390/cancers12092369] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022] Open
Abstract
Photodynamic therapy (PDT) is an attractive cancer treatment modality. Talaporfin sodium, a second-generation photosensitizer, results in lower systemic toxicity and relatively better selective tumor destruction than first-generation photosensitizers. However, the mechanism through which PDT induces vascular shutdown is unclear. In this study, the in vitro effects of talaporfin sodium-based PDT on human umbilical vein endothelial cells (HUVECs) were determined through cell viability and endothelial tube formation assays, and evaluation of the tubulin and F-actin dynamics and myosin light chain (MLC) phosphorylation. Additionally, the effects on tumor blood flow and tumor vessel destruction were assessed in vivo. In the HUVECs, talaporfin sodium-based PDT induced endothelial tube destruction and microtubule depolymerization, triggering the formation of F-actin stress fibers and a significant increase in MLC phosphorylation. However, pretreatment with the Rho-associated protein kinase (ROCK) inhibitor, Y27632, completely prevented PDT-induced stress fiber formation and MLC phosphorylation. The in vivo analysis and pathological examination revealed that the PDT had significantly decreased the tumor blood flow and the active area of the tumor vessel. We concluded that talaporfin sodium-based PDT induces the shutdown of existing tumor vessels via the RhoA/ROCK pathway by activating the Rho-GTP pathway and decreasing the tumor blood flow.
Collapse
Affiliation(s)
| | - Mamoru Tanaka
- Correspondence: ; Tel.: +81-52-853-8211; Fax: +81-52-852-0952
| | | | | | | | | |
Collapse
|
150
|
Santoro A, Bufo P, Russo G, Cagiano S, Papagerakis S, Bucci P, Aquino G, Longo F, Feola A, Giordano A, Di Carlo A, Di Domenico M, Pannone G. Expression and clinical implication of cyclooxygenase-2 and E-cadherin in oral squamous cell carcinomas. Cancer Biol Ther 2020. [PMID: 26218314 PMCID: PMC7537792 DOI: 10.1080/15384047.2015.1071741] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epithelial-Mesenchymal Transition (EMT) and angiogenesis are crucial events for development of aggressive and often fatal Oral Squamous Cell Carcinomas (OSCCs). Both promote cancer progression and metastasis development, but while the former induces the loss of E-cadherin expression and, hence cadherin switching; the latter produces hematic blood vessel neo-formation and contribute to OSCC cell growth, tumor mass development, and dissemination. Cyclooxygenase-2 (COX-2) has an important role, not only in angiogenic mechanisms, but also in favoring cancer invasion. Indeed it decreases the expression of E-cadherin and leads to phenotypic changes in epithelial cells (EMT) enhancing their carcinogenic potential. Our aim is to evaluate the interplay between E-cadherin cytoplasmic delocalization, COX-2 up-regulation and COX-2 induced neo-angiogenesis in 120 cases of OSCC. We have analyzed the distribution and the number of neo-formed endothelial buds surrounding infiltrating cells that express COX-2, as well as the neo-formed vessels in chronic inflammatory infiltrate, which surround the tumor. A double immunostaining method was employed in order to verify co-localization of endothelial cell marker (CD34) and COX-2. IHC has also been used to assess E-cadherin expression. Our data demonstrate that the OSCC cells, which lose membranous E-cadherin staining, acquiring a cytoplasmic delocalization, overexpress COX-2. Moreover, we find a new CD34+ vessel formation (sprouting angiogenesis). Only basaloid type of OSCC showes low level of COX-2 expression together with very low level of neo-angiogenesis and consequent tumor necrosis. The well-known anti-metastatic effect of certain COX-2 inhibitors suggests that these molecules might have clinical utility in the management of advanced cancers.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Anatomic Pathology, 'Giovanni Paolo II' Foundation-UCSC , Campobasso, Italy
| | - Pantaleo Bufo
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Giuseppe Russo
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Simona Cagiano
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Silvana Papagerakis
- Laboratory of Head and Neck Cancer Invasion and Metastasis, Department of Otolaryngology, Head and Neck Oncology, University of Michigan, Medical School , Ann Arbor, MI, USA
| | - Paolo Bucci
- Department of Neuroscience, Reproductive and Odontostomatologies Science, University of Naples 'Federico II' , Naples, Italy
| | - Gabriella Aquino
- Section of Pathological Anatomy, National Cancer Institute 'G. Pascale Foundation' , Naples, Italy
| | - Francesco Longo
- Head and Neck Medical Oncology Unit, National Cancer Institute 'G. Pascale Foundation' , Naples, Italy
| | - Antonia Feola
- Department of Biology, University of Naples , Naples, Italy.,Department of Biochemistry, Biophisic and General Pathology Second University of Naples , Naples, Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena , Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia, PA, USA
| | - Angelina Di Carlo
- Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome , Rome, Italy
| | - Marina Di Domenico
- Department of Biochemistry, Biophisic and General Pathology Second University of Naples , Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia, PA, USA
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| |
Collapse
|