1
|
Jayab NA, Abed A, Talaat IM, Hamoudi R. The molecular mechanism of NF-κB dysregulation across different subtypes of renal cell carcinoma. J Adv Res 2025; 72:501-514. [PMID: 39094893 DOI: 10.1016/j.jare.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The nuclear factor kappa B (NF-κB) is a critical pathway that regulates various cellular functions, including immune response, proliferation, growth, and apoptosis. Furthermore, this pathway is tightly regulated to ensure stability in the presence of immunogenic triggers or genotoxic stimuli. The lack of control of the NF-κB pathway can lead to the initiation of different diseases, mainly autoimmune diseases and cancer, including Renal cell carcinoma (RCC). RCC is the most common type of kidney cancer and is characterized by complex genetic composition and elusive molecular mechanisms. AIM OF REVIEW The current review summarizes the mechanism of NF-κB dysregulation in different subtypes of RCC and its impact on pathogenesis. KEY SCIENTIFIC CONCEPT OF REVIEW This review highlights the prominent role of NF-κB in RCC development and progression by driving the expression of multiple genes and interplaying with different pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. In silico analysis of RCC cohorts and molecular studies have revealed that multiple NF-κB members and target genes are dysregulated. The dysregulation includes receptors such as TLR2, signal-transmitting members including RelA, and target genes, for instance, HIF-1α. The lack of effective regulatory mechanisms results in a constitutively active NF-κB pathway, which promotes cancer growth, migration, and survival. In this review, we comprehensively summarize the role of dysregulated NF-κB-related genes in the most common subtypes of RCC, including clear cell RCC (ccRCC), chromophobe RCC (chRCC), and papillary RCC (PRCC).
Collapse
Affiliation(s)
- Nour Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Alaa Abed
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, 21131 Alexandria, Egypt.
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, 27272 Sharjah, United Arab Emirates; Division of Surgery and Interventional Science, University College London, London, United Kingdom; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
2
|
Purohit S, Mandal G, Biswas S, Dalui S, Gupta A, Chowdhury SR, Bhattacharyya A. AXL/GAS6 signaling governs differentiation of tumor-associated macrophages in breast cancer. Exp Cell Res 2025; 444:114324. [PMID: 39510154 DOI: 10.1016/j.yexcr.2024.114324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Most epithelial cancers are infiltrated by prognostically relevant myelomonocytic cells. Immunosuppressive tumor associated macrophages (TAMs) and their precursor monocytic myeloid-derived suppressor cells (MDSCs) have previously been associated with worse outcomes in human breast cancer (BCa), yet the mechanism of immunosuppressive TAMs-polarization from myelomonocytic precursors is not completely understood. In this study, we show that persuaded AXL/GAS6 pathway alters macrophage phenotype from HLA-DRhighCD206lowCD163low classical phagocytic into HLA-DRlowCD206highCD163high immunosuppressive ones with accelerated BCa progression, and increased angiogenesis signature and invasion ability of cancer cells at tumor beds. Notably, both AXL and GAS6 expressions are upregulated in human invasive breast carcinoma, with maximum expression in triple negative histology type. Mechanistically, we demonstrate that AXL/GAS6 signaling drives immunosuppression by governing increased immunosuppressive IL10 production while dampening IL-1β expression within the tumor microenvironment (TME) of BCa. Further, AXL/GAS6 signaling promotes angiogenesis through the activation of PI3K/AKT and NF-κB signaling pathways. Our results unveil role of AXL/GAS6 axis in the differentiation of TAMs, which governs malignant growth, and suggest that therapies that uncouple AXL/GAS6 axis may exhibit therapeutic opportunity for otherwise undruggable Triple Negative Breast Cancer (TNBC) patients.
Collapse
Affiliation(s)
- Suman Purohit
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Department of Zoology, Gurudas College, 1/1, Suren Sarkar Road, Phool Bagan, Kolkata, 700054, West Bengal, India
| | - Gunjan Mandal
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Division of Cancer Biology, DBT-Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Subir Biswas
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Tumor Immunology and Immunotherapy, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, Maharashtra, India
| | - Shauryabrota Dalui
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Arnab Gupta
- Department of Surgical Oncology, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata, 700063, West Bengal, India
| | - Sougata Roy Chowdhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Translational Immunology Laboratory, Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India.
| |
Collapse
|
3
|
Şancı E, Köksal Karayıldırım Ç, Dağdeviren M, Yiğittürk G, Buhur A, Erbaş O, Yavaşoğlu A, Karabay Yavaşoğlu NÜ. Oxidative stress and inflammatory markers in streptozotocin-induced acute and subacute toxicity response. Drug Chem Toxicol 2024; 47:933-948. [PMID: 38348650 DOI: 10.1080/01480545.2024.2315150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/01/2024] [Indexed: 11/21/2024]
Abstract
Streptozotocin (STZ) is used as a diabetes-inducing agent in experimental animal studies. However, it is known that STZ-induced diabetic animals show significant increases in oxidative stress parameters and neurodegeneration besides their blood glucose level. In this study, the acute and subacute toxic effects of STZ on the liver, sciatic nerve, and brain tissues were investigated in vivo rat model. Sprague-Dawley rats were divided into two groups; while 50 mg/kg STZ was administered ip to the STZ group, only saline was administered to the control group. After STZ administration, three units (100 U/mL) of subcutaneous insulin glargine were applied daily to prevent the formation of diabetes. At 24 h, 1,2, and 4 weeks after applications, rats from each group were sacrificed and tissues were removed under anesthesia. At the end of the study, compared to the control, a significant decrease in SOD and GST activity and an increase in lipid peroxidation were detected in the liver and sciatic tissues of rats in the STZ-treated group in the first 24h. Considering the TUNEL, NFκB, and NOS2 expressions, it was noted that while the effects of STZ on the liver were observed in the acute stage (24h), it had subacute effects on the brain. When apoptosis-related gene expression (Bcl-2, Bax, CASP3, CASP8, CASP9, TNF-α) and immunohistochemistry were evaluated, the apoptotic effect of STZ was observed mostly in sciatic nerve tissues. Within the scope of the study, it was revealed that STZ did not only show selective toxicity to pancreatic β cells but also very toxic to other tissues and organs.
Collapse
Affiliation(s)
- Ebru Şancı
- Center for Drug Development and Pharmacokinetic Applications, Ege University, Bornova Izmir, Turkey
| | | | | | - Gürkan Yiğittürk
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Aylin Buhur
- Department of Histology and Embryology, Ege University, Bornova Izmir, Turkey
| | - Oytun Erbaş
- Department of Physiology, Demiroğlu Bilim University, Istanbul, Turkey
| | - Altuğ Yavaşoğlu
- Department of Histology and Embryology, Ege University, Bornova Izmir, Turkey
| | - Nefise Ülkü Karabay Yavaşoğlu
- Center for Drug Development and Pharmacokinetic Applications, Ege University, Bornova Izmir, Turkey
- Department of Biology, Ege University, Bornova Izmir, Turkey
| |
Collapse
|
4
|
Liu BQ, Bai Y, Chen DP, Zhang YM, Wang TZ, Chen JR, Liu XY, Zheng B, Cui ZL. Intratumoural microorganism can affect the progression of hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:4232-4243. [DOI: 10.4251/wjgo.v16.i10.4232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Several recent studies have confirmed that intratumoural microorganisms can affect the occurrence and development of hepatocellular carcinoma (HCC); however, their role in tumor progression remains unclear. Hence, there is a need for further research on the role of intratumoural microorganisms in HCC.
AIM To investigate the changes in intratumoural microorganisms in HCC and the effect of Propionibacterium on HCC progression.
METHODS HCC and normal liver tissue specimens were subjected to fluorescence in situ hybridization (FISH). After performing 16S rRNA sequencing on HCC and peritumoral tissues to analyze the differences between the two groups. Propionibacterium was cocultured with HCC cells in vitro. Changes in cell proliferation and migration capacity were evaluated. The expression of NF-κB pathway related proteins in tumor cells was compared. The orthotopic liver implantation model and the subcutaneous xenograft model were constructed. liver tissues and subcutaneous tumors were collected 2 weeks later.
RESULTS FISH demonstrated the presence of microorganisms in HCC and normal liver tissues. 16S rRNA sequencing revealed an abundance of Lysobacter, Lachnospiraceae, Pseudomonas, and Lactobacillus in HCC tissues. The distribution and abundance of Propionibacterium showed differences between HCC and peritumoral tissues (P < 0.05). In vitro studies demonstrated that Propionibacterium and its metabolite propionic acid (PA) inhibited the proliferation and migration of HCC cells (P < 0.05). The expression of the proteins in NF-κB signaling pathway also decreased in HCC cells (P < 0.05).
CONCLUSION Microorganisms in HCC and normal liver tissues displayed significant disparities. The PA-producing bacterium Propionibacterium in HCC exerts an effect on the NF-κB pathway, thereby affecting the biological behavior of HCC.
Collapse
Affiliation(s)
- Bao-Qun Liu
- First Central Clinical College, Tianjin Medical University, Tianjin 300070, China
| | - Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| | - Da-Peng Chen
- First Central Clinical College, Tianjin Medical University, Tianjin 300070, China
| | - Ya-Min Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| | - Tian-Ze Wang
- Tianjin First Central Hospital Clinic Institute, School of Medicine, Nankai University, Tianjin 300192, China
| | - Jing-Rui Chen
- First Central Clinical College, Tianjin Medical University, Tianjin 300070, China
| | - Xiang-Yu Liu
- Tianjin First Central Hospital Clinic Institute, School of Medicine, Nankai University, Tianjin 300192, China
| | - Bin Zheng
- School of Medicine, Tianjin University, Tianjin 300072, China
| | - Zi-Lin Cui
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| |
Collapse
|
5
|
Koychev I, Reid G, Nguyen M, Mentz RJ, Joyce D, Shah SH, Holman RR. Inflammatory proteins associated with Alzheimer's disease reduced by a GLP1 receptor agonist: a post hoc analysis of the EXSCEL randomized placebo controlled trial. Alzheimers Res Ther 2024; 16:212. [PMID: 39358806 PMCID: PMC11448378 DOI: 10.1186/s13195-024-01573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists are a viable option for the prevention of Alzheimer's disease (AD) but the mechanisms of this potential disease modifying action are unclear. We investigated the effects of once-weekly exenatide (EQW) on AD associated proteomic clusters. METHODS The Exenatide Study of Cardiovascular Event Lowering study compared the cardiovascular effects of EQW 2 mg with placebo in 13,752 people with type 2 diabetes mellitus. 4,979 proteins were measured (Somascan V0.4) on baseline and 1-year plasma samples of 3,973 participants. C-reactive protein (CRP), ficolin-2 (FCN2), plasminogen activator inhibitor 1 (PAI-1), soluble vascular cell adhesion protein 1 (sVCAM1) and 4 protein clusters were tested in multivariable mixed models. RESULTS EQW affected FCN2 (Cohen's d -0.019), PAI-1 (Cohen's d -0.033), sVCAM-1 (Cohen's d 0.035) and a cytokine-cytokine cluster (Cohen's d 0.037) significantly compared with placebo. These effects were sustained in individuals over the age of 65 but not in those under 65. CONCLUSIONS EQW treatment was associated with significant change in inflammatory proteins associated with AD. TRIAL REGISTRATION EXSCEL is registered on ClinicalTrials.gov: NCT01144338 on 10th of June 2010.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK.
| | - Graham Reid
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Maggie Nguyen
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
| | | | - Dan Joyce
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Svati H Shah
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Diaz LC, Silva PGDB, Dantas TS, Mota MRL, Alves APNN, Rodrigues MIDQ, Mesquita KC, Filho OVDO, Sousa FB. Naltrexone accelerated oral traumatic ulcer healing and downregulated TLR-4/NF-kB pathway in Wistar rats. Arch Oral Biol 2024; 166:106047. [PMID: 39024694 DOI: 10.1016/j.archoralbio.2024.106047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE To assess the effect of naltrexone on oral mucosal healing using a traumatic ulcer model DESIGN: Wistar rats (n = 112) received distilled water (control) or naltrexone (0.5, 10, or 50 mg/kg/day). Ulcers were induced on the buccal mucosa using a round skin biopsy punch (diameter 6 mm). Euthanasia was performed on days 1, 3, 7, and 14. Healing was assessed by ulcer area, histological scores, histomorphometric analysis (number of polymorphonuclears, mononuclears, and fibroblasts), and collagen percentage. Immunohistochemistry for TLR-2, TLR-4, NF-kB, and CD31 was evaluated. Nociceptive threshold was measured daily. RESULTS The 50 mg/kg group showed reduced ulcer area on days 1 (p < 0.001), 3 (p < 0.05), and 14 (p < 0.01). In this group, there was, on day 14, an increase in the percentage of reepithelization (p = 0.043) and collagen (p < 0.05), an increase in connective tissue maturation (p = 0.016), and on day 7 an increase in fibroblasts (p < 0.001). The 10 mg/kg dose reduced the ulcer area on day 1 (p < 0.001). The 50 mg/kg group showed lower expression of TLR-4 (p < 0.001) on day 1, NF-kB on days 1 (p < 0.05) and 14 (p < 0.05), and CD31 on day 14 (p < 0.05). The 0.5 and 10 mg/kg doses reduced TLR-4 expression on day 1 (p < 0.05; p < 0.01, respectively). Nociceptive threshold increased in the 50 mg/kg group (p < 0.01). CONCLUSION Naltrexone enhanced traumatic oral ulcer healing by reducing TLR-4/NF-kB signaling and promoting fibroblast proliferation and collagen deposition. Additionally, naltrexone reduced pain in rats.
Collapse
Affiliation(s)
- Liova Chabot Diaz
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil.
| | - Paulo Goberlânio de Barros Silva
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil.
| | | | - Mário Rogério Lima Mota
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil.
| | - Ana Paula Negreiros Nunes Alves
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil.
| | - Maria Imaculada de Queiroz Rodrigues
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil.
| | | | - Osias Vieira de Oliveira Filho
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil.
| | - Fabrício Bitu Sousa
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, School of Pharmacy, Dentistry and Nursing Federal University of Ceara, Fortaleza, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil.
| |
Collapse
|
7
|
Cai Z, Gao L, Hu K, Wang QM. Parthenolide enhances the metronomic chemotherapy effect of cyclophosphamide in lung cancer by inhibiting the NF-kB signaling pathway. World J Clin Oncol 2024; 15:895-907. [PMID: 39071467 PMCID: PMC11271733 DOI: 10.5306/wjco.v15.i7.895] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/11/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Parthenolide (PTL), a sesquiterpene lactone derived from the medicinal herb Chrysanthemum parthenium, exhibits various biological effects by targeting NF-kB, STAT3, and other pathways. It has emerged as a promising adjunct therapy for multiple malignancies. AIM To evaluate the in vitro and in vivo effect of PTL on cyclophosphamide (CTX) metronomic chemotherapy. METHODS The cytotoxicity of PTL and CTX on Lewis lung cancer cells (LLC cells) was assessed by measuring cell activity and apoptosis. The anti-tumor efficiency was evaluated using a tumor xenograft mice model, and the survival of mice and tumor volume were monitored. Additionally, the collected tumor tissues were analyzed for tumor microenvironment indicators and inflammatory factors. RESULTS In vitro, PTL demonstrated a synergistic effect with CTX in inhibiting the growth of LLC cells and promoting apoptosis. In vivo, metronomic chemotherapy combined with PTL and CTX improved the survival rate of tumor-bearing mice and reduced tumor growth rate. Furthermore, metronomic chemotherapy combined with PTL and CTX reduced NF-κB activation and improved the tumor immune microenvironment by decreasing tumor angiogenesis, reducing Transforming growth factor β, and α-SMA positive cells. CONCLUSION PTL is an efficient compound that enhances the metronomic chemotherapy effects of CTX both in vitro and in vivo, suggesting its potential as a supplementary therapeutic strategy in metronomic chemotherapy to improve the chemotherapy effects.
Collapse
Affiliation(s)
- Zheng Cai
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, Henan Province, China
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Lang Gao
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Kai Hu
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Qi-Ming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, Henan Province, China
| |
Collapse
|
8
|
Kim NY, Park HM, Park JY, Kim U, Shin HY, Lee HP, Hong JT, Yoon DY. MMPP is a novel VEGFR2 inhibitor that suppresses angiogenesis via VEGFR2/AKT/ERK/NF-κB pathway. BMB Rep 2024; 57:244-249. [PMID: 37964635 PMCID: PMC11139683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/17/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023] Open
Abstract
Many types of cancer are associated with excessive angiogenesis. Anti-angiogenic treatment is an effective strategy for treating solid cancers. This study aimed to demonstrate the inhibitory effects of (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP) in VEGFA-induced angiogenesis. The results indicated that MMPP effectively suppressed various angiogenic processes, such as cell migration, invasion, tube formation, and sprouting of new vessels in human umbilical vein endothelial cells (HUVECs) and mouse aortic ring. The inhibitory mechanism of MMPP on angiogenesis involves targeting VEGFR2. MMPP showed high binding affinity for the VEGFR2 ATP-binding domain. Additionally, MMPP improved VEGFR2 thermal stability and inhibited VEGFR2 kinase activity, suppressing the downstream VEGFR2/AKT/ERK pathway. MMPP attenuated the activation and nuclear translocation of NF-κB, and it downregulated NF-κB target genes such as VEGFA, VEGFR2, MMP2, and MMP9. Furthermore, conditioned medium from MMPP-treated breast cancer cells effectively inhibited angiogenesis in endothelial cells. These results suggested that MMPP had great promise as a novel VEGFR2 inhibitor with potent anti-angiogenic properties for cancer treatment via VEGFR2/AKT/ERK/NF-κB signaling pathway. [BMB Reports 2024; 57(5): 244-249].
Collapse
Affiliation(s)
- Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Uijin Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Korea
| | - Ha Youn Shin
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
9
|
Kim NY, Park HM, Park JY, Kim U, Shin HY, Lee HP, Hong JT, Yoon DY. MMPP is a novel VEGFR2 inhibitor that suppresses angiogenesis via VEGFR2/AKT/ERK/NF-κB pathway. BMB Rep 2024; 57:244-249. [PMID: 37964635 PMCID: PMC11139683 DOI: 10.5483/bmbrep.2023-0150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/17/2023] [Accepted: 11/03/2023] [Indexed: 12/26/2024] Open
Abstract
Many types of cancer are associated with excessive angiogenesis. Anti-angiogenic treatment is an effective strategy for treating solid cancers. This study aimed to demonstrate the inhibitory effects of (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP) in VEGFA-induced angiogenesis. The results indicated that MMPP effectively suppressed various angiogenic processes, such as cell migration, invasion, tube formation, and sprouting of new vessels in human umbilical vein endothelial cells (HUVECs) and mouse aortic ring. The inhibitory mechanism of MMPP on angiogenesis involves targeting VEGFR2. MMPP showed high binding affinity for the VEGFR2 ATP-binding domain. Additionally, MMPP improved VEGFR2 thermal stability and inhibited VEGFR2 kinase activity, suppressing the downstream VEGFR2/AKT/ERK pathway. MMPP attenuated the activation and nuclear translocation of NF-κB, and it downregulated NF-κB target genes such as VEGFA, VEGFR2, MMP2, and MMP9. Furthermore, conditioned medium from MMPP-treated breast cancer cells effectively inhibited angiogenesis in endothelial cells. These results suggested that MMPP had great promise as a novel VEGFR2 inhibitor with potent anti-angiogenic properties for cancer treatment via VEGFR2/AKT/ERK/NF-κB signaling pathway. [BMB Reports 2024; 57(5): 244-249].
Collapse
Affiliation(s)
- Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Uijin Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Korea
| | - Ha Youn Shin
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
10
|
Chakraborty A, Ghosh S, Chakraborty MP, Mukherjee S, Roy SS, Das R, Acharya M, Mukherjee A. Inhibition of NF-κB-Mediated Proinflammatory Transcription by Ru(II) Complexes of Anti-Angiogenic Ligands in Triple-Negative Breast Cancer. J Med Chem 2024; 67:5902-5923. [PMID: 38520399 DOI: 10.1021/acs.jmedchem.4c00169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
Nuclear factor kappa beta (NF-κB) plays a pivotal role in breast cancer, particularly triple-negative breast cancer, by promoting inflammation, proliferation, epithelial-mesenchymal transition, metastasis, and drug resistance. Upregulation of NF-κB boosts vascular endothelial growth factor (VEGF) expression, assisting angiogenesis. The Ru(II) complexes of methyl- and dimethylpyrazolyl-benzimidazole N,N donors inhibit phosphorylation of ser536 in p65 and translocation of the NF-κB heterodimer (p50/p65) to the nucleus, disabling transcription to upregulate inflammatory signaling. The methyl- and dimethylpyrazolyl-benzimidazole inhibit VEGFR2 phosphorylation at Y1175, disrupting downstream signaling through PLC-γ and ERK1/2, ultimately suppressing Ca(II)-signaling. Partial release of the antiangiogenic ligand in a reactive oxygen species-rich environment is possible as per our observation to inhibit both NF-κB and VEGFR2 by the complexes. The complexes are nontoxic to zebrafish embryos up to 50 μM, but the ligands show strong in vivo antiangiogenic activity at 3 μM during embryonic growth in Tg(fli1:GFP) zebrafish but no visible effect on the adult phase.
Collapse
Affiliation(s)
- Ayan Chakraborty
- Department of Chemical Sciences and Centre for Advanced Functional Materials (CAFM), Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur 741246, India
| | - Shilpendu Ghosh
- Department of Chemical Sciences and Centre for Advanced Functional Materials (CAFM), Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur 741246, India
| | - Manas Pratim Chakraborty
- Department of Biological Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur Campus, Mohanpur 741246, India
| | - Sujato Mukherjee
- Department of Chemical Sciences and Centre for Advanced Functional Materials (CAFM), Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur 741246, India
| | | | - Rahul Das
- Department of Biological Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur Campus, Mohanpur 741246, India
| | | | - Arindam Mukherjee
- Department of Chemical Sciences and Centre for Advanced Functional Materials (CAFM), Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur 741246, India
| |
Collapse
|
11
|
Jacob JR, Singh R, Okamoto M, Chakravarti A, Palanichamy K. miRNA-194-3p represses NF-κB in gliomas to attenuate iPSC genes and proneural to mesenchymal transition. iScience 2024; 27:108650. [PMID: 38226170 PMCID: PMC10788216 DOI: 10.1016/j.isci.2023.108650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/01/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024] Open
Abstract
Severe tumor heterogeneity drives the aggressive and treatment refractory nature of glioblastomas (GBMs). While limiting GBM heterogeneity offers promising therapeutic potential, the underlying mechanisms that regulate GBM plasticity remain poorly understood. We utilized 14 patient-derived and four commercially available cell lines to uncover miR-194-3p as a key epigenetic determinant of stemness and transcriptional subtype in GBM. We demonstrate that miR-194-3p degrades TAB2, an important mediator of NF-κB activity, decreasing NF-κB transcriptional activity. The loss in NF-κB activity following miR-194-3p overexpression or TAB2 silencing decreased expression of induced pluripotent stem cell (iPSC) genes, inhibited the oncogenic IL-6/STAT3 signaling axis, suppressed the mesenchymal transcriptional subtype in relation to the proneural subtype, and induced differentiation from the glioma stem cell (GSC) to monolayer (ML) phenotype. miR-194-3p/TAB2/NF-κB signaling axis acts as an epigenetic switch that regulates GBM plasticity and targeting this signaling axis represents a potential strategy to limit transcriptional heterogeneity in GBMs.
Collapse
Affiliation(s)
- John Ryan Jacob
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rajbir Singh
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Masa Okamoto
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
12
|
Ahmed SA, Mendonca P, Messeha SS, Soliman KFA. Anticancer Effects of Fucoxanthin through Cell Cycle Arrest, Apoptosis Induction, and Angiogenesis Inhibition in Triple-Negative Breast Cancer Cells. Molecules 2023; 28:6536. [PMID: 37764312 PMCID: PMC10535858 DOI: 10.3390/molecules28186536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The absence of progesterone receptors, estrogen receptors, and human epidermal growth factor receptor-2 restricts the therapy choices for treating triple-negative breast cancer (TNBC). Moreover, conventional medication is not highly effective in treating TNBC, and developing effective therapeutic agents from natural bioactive compounds is a viable option. In this study, the anticancer effects of the natural compound fucoxanthin were investigated in two genetically different models of TNBC cells: MDA-MB-231 and MDA-MB-468 cells. Fucoxanthin had a significant anticancer effect in both cell lines at a concentration range of 1.56-300 µM. The compound decreased cell viability in both cell lines with higher potency in MDA-MB-468 cells. Meanwhile, proliferation assays showed similar antiproliferative effects in both cell lines after 48 h and 72 h treatment periods. Flow cytometry and Annexin V-FITC apoptosis assay revealed the ability of fucoxanthin to induce apoptosis in MDA-MB-231 only. Cell cycle arrest analysis showed that the compound also induced cell cycle arrest at the G1 phase in both cell lines, accompanied by more cell cycle arrest in MDA-MB-231 cells at S-phase and a higher cell cycle arrest in the MDA-MB-468 cells at G2-phase. Wound healing and migration assay showed that in both cell lines, fucoxanthin prevented migration, but was more effective in MDA-MB-231 cells in a shorter time. In both angiogenic cytokine array and RT-PCR studies, fucoxanthin (6.25 µM) downregulated VEGF-A and -C expression in TNF-α-stimulated (50 ng/mL) MDA-MB-231, but not in MDA-MB-468 cells on the transcription and protein levels. In conclusion, this study shows that fucoxanthin was more effective in MDA-MB-231 TNBC cells, where it can target VEGF-A and VEGF-C, inhibit cell proliferation and cell migration, and induce cell cycle arrest and apoptosis-the most crucial cellular processes involved in breast cancer development and progression.
Collapse
Affiliation(s)
- Shade’ A. Ahmed
- Division of Pharmaceutical Sciences, Institute of Public Health, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Samia S. Messeha
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, Institute of Public Health, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| |
Collapse
|
13
|
A’yuni DQ, Sa’adi A, Widjiati W. Ethanol extract of basil ( Ocimum Basilicum L.) leaves inhibits endometriosis growth in a mouse model by modulating vascular endothelial growth factor (VEGF) expression. J Med Life 2023; 16:1224-1230. [PMID: 38024822 PMCID: PMC10652688 DOI: 10.25122/jml-2023-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/29/2023] [Indexed: 12/01/2023] Open
Abstract
The objective of this study was to examine the effect of administering an ethanol extract obtained from basil leaves on the expression of vascular endothelial growth factor (VEGF) and the severity of endometriosis lesions in a mouse model. A total of 28 female mice, aged 2-3 months and weighing 20-30 grams, were randomly divided into four groups: the control group (C), treatment group 1 (T1) receiving a dose of basil leaf ethanol extract (0.21 mg/g-BW), treatment group 2 (T2) receiving a higher dose (0.42 mg/g BW), and treatment group 3 (T3) receiving the highest dose (0.84 mg/g-BW). Each group underwent a 14-day treatment period, and tissue samples were collected on the 29th day. An immunohistochemical examination was conducted to assess the expression of VEGF and evaluate the severity of endometriosis lesions. The statistical analysis of VEGF expression revealed a significant difference (p=0.026; p<0.05), with the most pronounced effects observed when administering basil leaf ethanol extract at doses of 0.21 mg/g-BW and 0.42 mg/g-BW. Although not statistically significant (p=0.271; p<0.05), a reduction in the severity of endometriosis lesions was observed following the administration of basil leaf ethanol extract at doses of 0.21 mg/g-BW and 0.42 mg/g-BW. Administering basil leaf ethanol extract at doses of 0.21 mg/g-BW and 0.42 mg/g-BW effectively decreased VEGF expression and limited the severity of endometriosis lesions.
Collapse
Affiliation(s)
| | - Ashon Sa’adi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Widjiati Widjiati
- Department of Veterinary Science, Faculty of Veterinary Medicine, Airlangga University, Surabaya, Indonesia
| |
Collapse
|
14
|
Namjoo M, Ghafouri H, Assareh E, Aref AR, Mostafavi E, Hamrahi Mohsen A, Balalaie S, Broussy S, Asghari SM. A VEGFB-Based Peptidomimetic Inhibits VEGFR2-Mediated PI3K/Akt/mTOR and PLCγ/ERK Signaling and Elicits Apoptotic, Antiangiogenic, and Antitumor Activities. Pharmaceuticals (Basel) 2023; 16:906. [PMID: 37375853 DOI: 10.3390/ph16060906] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) mediates VEGFA signaling mainly through the PI3K/AKT/mTOR and PLCγ/ERK1/2 pathways. Here we unveil a peptidomimetic (VGB3) based on the interaction between VEGFB and VEGFR1 that unexpectedly binds and neutralizes VEGFR2. Investigation of the cyclic and linear structures of VGB3 (named C-VGB3 and L-VGB3, respectively) using receptor binding and cell proliferation assays, molecular docking, and evaluation of antiangiogenic and antitumor activities in the 4T1 mouse mammary carcinoma tumor (MCT) model showed that loop formation is essential for peptide functionality. C-VGB3 inhibited proliferation and tubulogenesis of human umbilical vein endothelial cells (HUVECs), accounting for the abrogation of VEGFR2, p-VEGFR2 and, subsequently, PI3K/AKT/mTOR and PLCγ/ERK1/2 pathways. In 4T1 MCT cells, C-VGB3 inhibited cell proliferation, VEGFR2 expression and phosphorylation, the PI3K/AKT/mTOR pathway, FAK/Paxillin, and the epithelial-to-mesenchymal transition cascade. The apoptotic effects of C-VGB3 on HUVE and 4T1 MCT cells were inferred from annexin-PI and TUNEL staining and activation of P53, caspase-3, caspase-7, and PARP1, which mechanistically occurred through the intrinsic pathway mediated by Bcl2 family members, cytochrome c, Apaf-1 and caspase-9, and extrinsic pathway via death receptors and caspase-8. These data indicate that binding regions shared by VEGF family members may be important in developing novel pan-VEGFR inhibitors that are highly relevant in the pathogenesis of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Mohadeseh Namjoo
- Department of Biology, University Campus II, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Hossein Ghafouri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Elham Assareh
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ali Hamrahi Mohsen
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran P.O. Box 1841, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, Tehran P.O. Box 1841, Iran
| | - Sylvain Broussy
- CiTCoM, UMR CNRS 8038, U1268 INSERM, UFR de Pharmacie, Faculté de Santé, Université Paris Cité, 75006 Paris, France
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran P.O. Box 1841, Iran
| |
Collapse
|
15
|
Srinath A, Xie B, Li Y, Sone JY, Romanos S, Chen C, Sharma A, Polster S, Dorrestein PC, Weldon KC, DeBiasse D, Moore T, Lightle R, Koskimäki J, Zhang D, Stadnik A, Piedad K, Hagan M, Shkoukani A, Carrión-Penagos J, Bi D, Shen L, Shenkar R, Ji Y, Sidebottom A, Pamer E, Gilbert JA, Kahn ML, D'Souza M, Sulakhe D, Awad IA, Girard R. Plasma metabolites with mechanistic and clinical links to the neurovascular disease cavernous angioma. COMMUNICATIONS MEDICINE 2023; 3:35. [PMID: 36869161 PMCID: PMC9984539 DOI: 10.1038/s43856-023-00265-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Cavernous angiomas (CAs) affect 0.5% of the population, predisposing to serious neurologic sequelae from brain bleeding. A leaky gut epithelium associated with a permissive gut microbiome, was identified in patients who develop CAs, favoring lipid polysaccharide producing bacterial species. Micro-ribonucleic acids along with plasma levels of proteins reflecting angiogenesis and inflammation were also previously correlated with CA and CA with symptomatic hemorrhage. METHODS The plasma metabolome of CA patients and CA patients with symptomatic hemorrhage was assessed using liquid-chromatography mass spectrometry. Differential metabolites were identified using partial least squares-discriminant analysis (p < 0.05, FDR corrected). Interactions between these metabolites and the previously established CA transcriptome, microbiome, and differential proteins were queried for mechanistic relevance. Differential metabolites in CA patients with symptomatic hemorrhage were then validated in an independent, propensity matched cohort. A machine learning-implemented, Bayesian approach was used to integrate proteins, micro-RNAs and metabolites to develop a diagnostic model for CA patients with symptomatic hemorrhage. RESULTS Here we identify plasma metabolites, including cholic acid and hypoxanthine distinguishing CA patients, while arachidonic and linoleic acids distinguish those with symptomatic hemorrhage. Plasma metabolites are linked to the permissive microbiome genes, and to previously implicated disease mechanisms. The metabolites distinguishing CA with symptomatic hemorrhage are validated in an independent propensity-matched cohort, and their integration, along with levels of circulating miRNAs, enhance the performance of plasma protein biomarkers (up to 85% sensitivity and 80% specificity). CONCLUSIONS Plasma metabolites reflect CAs and their hemorrhagic activity. A model of their multiomic integration is applicable to other pathologies.
Collapse
Affiliation(s)
- Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Bingqing Xie
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Ying Li
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, 150001, Harbin, Heilongjiang, China
| | - Je Yeong Sone
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Chang Chen
- Bioinformatics Core, Center for Research Informatics, The University of Chicago, Chicago, IL, 60637, USA
| | - Anukriti Sharma
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Pieter C Dorrestein
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Department of Pharmacology, The University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Kelly C Weldon
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Kristina Piedad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Matthew Hagan
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Abdallah Shkoukani
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Dehua Bi
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Le Shen
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Ashley Sidebottom
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Eric Pamer
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Mark D'Souza
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Dinanath Sulakhe
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA.
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| |
Collapse
|
16
|
Candesartan protects against unilateral peripheral limb ischemia in type-2 diabetic rats: Possible contribution of PI3K-Akt-eNOS-VEGF angiogenic signaling pathway. Int Immunopharmacol 2023; 116:109817. [PMID: 36773570 DOI: 10.1016/j.intimp.2023.109817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
Type-2 diabetes (T2DM) is known to be highly associated with increased risk for vascular complications including peripheral arterial diseases (PAD). Critical limb ischemia (CLI) is the most advanced stage of PAD. Current therapeutic options for diabetic patients experiencing vascular complications are limited to surgical revascularization with no effective pharmacotherapy available for clinical settings. This study is dedicated to evaluate the angiogenic potential of candesartan an angiotensin-II receptor blocker in an experimental model of vascular complications associating T2DM. T2DM was induced in rats through feeding with high fat diet for 6 weeks, followed by injection with streptozotocin (STZ, 30 mg/kg; i.p). After establishment of T2DM, unilateral CLI was induced through the ligation and excision of superficial femoral artery. Candesartan treatment (10 or 30 mg/kg; orally) was initiated one day post CLI and thereafter once daily for up to 14 days. T2DM rats that underwent CLI demonstrated impaired angiogenic signaling, increased inflammation and apoptosis in gastrocnemius muscle (GC). Candesartan reversed ischemic insult in T2DM rats subjected to unilateral CLI and induced reparative angiogenesis that was evident by increase in p-PI3K/PI3K, p-Akt/Akt, p-eNOS/eNOS, p-VEGFR2/VEGFR2 ratios, and VEGF levels. Candesartan treatment also increased levels of HO-1; while decreased caspase-3 apoptotic marker and levels of inflammatory markers; NF-κB and TNF-α, all of which were accompanied by preserved histological manifestations of GC muscles. Candesartan was able to combat limb ischemia under diabetic conditions which could pave the way for its therapeutic utility for diabetic patients experiencing vascular complications in clinical setting.
Collapse
|
17
|
Kandjani BZ, Hesari FS, Babaei E. Gemini curcumin inhibits 4T1 cancer cell proliferation and modulates the expression of apoptotic and metastatic genes in Balb/c mice model. Pathol Res Pract 2023; 243:154344. [PMID: 36738519 DOI: 10.1016/j.prp.2023.154344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/15/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND Despite the attractive anti-cancer effects, poor solubility and low bioavailability have restricted the clinical application of Curcumin. Recent findings show that Gemini nano-curcumin (Gemini-Cur) significantly improves the cellular uptake of Curcumin and its anti-cancer effect in tumor cells. Here, we aimed to assess the suppressive effect of Gemini-Cur on 4T1 breast cancer cells in vitro and, subsequently, in BALB/c mouse models. MATERIALS AND METHODS Fluorescence microscopy was employed to visualize cellular uptake and morphological changes of 4T1 cells during treatment with Gemini-Cur and void curcumin. MTT and annexin V/FITC assays were performed to study the toxic effect of Gemini-Cur on mouse cancer cells. For in vivo studies, BALB/c tumor-bearing mice were used to evaluate the inhibitory effect of Gemini-Cur in comparison with mice receiving free Curcumin and nanoparticles. RESULTS Our data showed that Gemini-Cur enters the cells and inhibits proliferation in a time- and dose-dependent manner. Annexin V/FITC confirmed apoptotic effect on 4T1 cells. In vivo studies also illustrated that tumor growth is suppressed in Gemini-Cur treated mice rather than controls. Expression studies demonstrated the modulation of apoptotic and metastatic genes, including Bax, Bcl-2, MMP-9, VEGF, and COX-2 in treated mice. CONCLUSION In conclusion, these data demonstrate the promising anti-cancer properties of Gemini-Cur on mice models. However, further studies at molecular and cellular levels are required to conclude this therapeutic advantage.
Collapse
Affiliation(s)
- Behzad Zaker Kandjani
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Esmaeil Babaei
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Interfaculty Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
18
|
Peng X, Zhang R, Zhang Y, Cai C. Nuclear Factor-Kappa B-induced miRNA-518a-5p represses trophoblast cell migration and invasion by the Nuclear Factor-Kappa B pathway. AN ACAD BRAS CIENC 2023; 95:e20220596. [PMID: 37132750 DOI: 10.1590/0001-3765202320220596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/03/2022] [Indexed: 05/04/2023] Open
Abstract
Preeclampsia is associated with the insufficient invasion of trophoblasts. NF-κB is a transcription factor in almost all mammalian cells and has been validated to be upregulated in the maternal circulation and placenta of women with preeclampsia. MiR-518a-5p is also overexpressed in pre-eclamptic placenta. The present study was designed to explore whether NF-κB can transcriptionally activate miR-518a-5p and investigate the influences of miR-518a-5p on the viability, apoptosis, migration, and invasion of HTR8/SVneo trophoblast. In situ hybridization and real time polymerase chain reaction were used to reveal miR-518a-5p expression in placenta tissues and HTR8/SVneo cells, respectively. Cell migration and invasion were detected using Transwell inserts. Our findings indicated that NF-κB p52, p50, and p65 can bind to miR-518a-5p gene promoter. MiR-518a-5p further influences the levels of p50 and p65 but not p52. HTR8/SVneo cell viability and apoptosis were not influenced by miR-518a-5p. However, miR-518a-5p represses the migratory/invasive capacities of HTR8/SVneo cell and decreased gelatinolytic activity of MMP2 and MMP9, which was reversed by an NF-κB inhibitor. To sum up, miR-518a-5p is induced by NF-κB and represses trophoblast cell migration and invasion by the NF-κB pathway.
Collapse
Affiliation(s)
- Xing Peng
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| | - Ruirui Zhang
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| | - Yumei Zhang
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| | - Chunyan Cai
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| |
Collapse
|
19
|
PARP Inhibitor Inhibits the Vasculogenic Mimicry through a NF-κB-PTX3 Axis Signaling in Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms232416171. [PMID: 36555812 PMCID: PMC9785325 DOI: 10.3390/ijms232416171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) are targeted therapies that inhibit PARP proteins which are involved in a variety of cell functions. PARPi may act as modulators of angiogenesis; however, the relationship between PARPi and the vasculogenic mimicry (VM) in breast cancer remains unclear. To determine whether PARPi regulate the vascular channel formation, we assessed whether the treatment with olaparib, talazoparib and veliparib inhibits the vascular channel formation by breast cancer cell lines. Here, we found that PARPi act as potent inhibitors of the VM formation in triple negative breast cancer cells, independently of the BRCA status. Mechanistically, we find that PARPi trigger and inhibit the NF-κB signaling, leading to the inhibition of the VM. We further show that PARPi decrease the expression of the angiogenic factor PTX3. Moreover, PTX3 rescued the PARPi-inhibited VM inhibition. In conclusion, our results indicate that PARPi, by targeting the VM, may provide a new therapeutic approach for triple negative breast cancer.
Collapse
|
20
|
Ghoneum A, Gonzalez D, Afify H, Shu J, Hegarty A, Adisa J, Kelly M, Lentz S, Salsbury F, Said N. Compound C Inhibits Ovarian Cancer Progression via PI3K-AKT-mTOR-NFκB Pathway. Cancers (Basel) 2022; 14:5099. [PMID: 36291886 PMCID: PMC9600774 DOI: 10.3390/cancers14205099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 12/04/2022] Open
Abstract
Epithelial Ovarian cancer (OvCa) is the leading cause of death from gynecologic malignancies in the United States, with most patients diagnosed at late stages. High-grade serous cancer (HGSC) is the most common and lethal subtype. Despite aggressive surgical debulking and chemotherapy, recurrence of chemo-resistant disease occurs in ~80% of patients. Thus, developing therapeutics that not only targets OvCa cell survival, but also target their interactions within their unique peritoneal tumor microenvironment (TME) is warranted. Herein, we report therapeutic efficacy of compound C (also known as dorsomorphin) with a novel mechanism of action in OvCa. We found that CC not only inhibited OvCa growth and invasiveness, but also blunted their reciprocal crosstalk with macrophages, and mesothelial cells. Mechanistic studies indicated that compound C exerts its effects on OvCa cells through inhibition of PI3K-AKT-NFκB pathways, whereas in macrophages and mesothelial cells, CC inhibited cancer-cell-induced canonical NFκB activation. We further validated the specificity of the PI3K-AKT-NFκB as targets of compound C by overexpression of constitutively active subunits as well as computational modeling. In addition, real-time monitoring of OvCa cellular bioenergetics revealed that compound C inhibits ATP production, mitochondrial respiration, and non-mitochondrial oxygen consumption. Importantly, compound C significantly decreased tumor burden of OvCa xenografts in nude mice and increased their sensitivity to cisplatin-treatment. Moreover, compound C re-sensitized patient-derived resistant cells to cisplatin. Together, our findings highlight compound C as a potent multi-faceted therapeutic in OvCa.
Collapse
Affiliation(s)
- Alia Ghoneum
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Daniela Gonzalez
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Hesham Afify
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Junjun Shu
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Abigail Hegarty
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Jemima Adisa
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Michael Kelly
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Comprehensive Cancer Center, Wake Forest Baptist Health Sciences, Winston Salem, NC 27157, USA
| | - Samuel Lentz
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Comprehensive Cancer Center, Wake Forest Baptist Health Sciences, Winston Salem, NC 27157, USA
- Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Freddie Salsbury
- Comprehensive Cancer Center, Wake Forest Baptist Health Sciences, Winston Salem, NC 27157, USA
- Department of Physics, Wake Forest University, Winston Salem, NC 27109, USA
| | - Neveen Said
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Comprehensive Cancer Center, Wake Forest Baptist Health Sciences, Winston Salem, NC 27157, USA
- Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
21
|
Liu P, Li Y, Wang W, Bai Y, Jia H, Yuan Z, Yang Z. Role and mechanisms of the NF-ĸB signaling pathway in various developmental processes. Biomed Pharmacother 2022; 153:113513. [DOI: 10.1016/j.biopha.2022.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022] Open
|
22
|
Casas BS, Arancibia-Altamirano D, Acevedo-La Rosa F, Garrido-Jara D, Maksaev V, Pérez-Monje D, Palma V. It takes two to tango: Widening our understanding of the onset of schizophrenia from a neuro-angiogenic perspective. Front Cell Dev Biol 2022; 10:946706. [PMID: 36092733 PMCID: PMC9448889 DOI: 10.3389/fcell.2022.946706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic debilitating mental disorder characterized by perturbations in thinking, perception, and behavior, along with brain connectivity deficiencies, neurotransmitter dysfunctions, and loss of gray brain matter. To date, schizophrenia has no cure and pharmacological treatments are only partially efficacious, with about 30% of patients describing little to no improvement after treatment. As in most neurological disorders, the main descriptions of schizophrenia physiopathology have been focused on neural network deficiencies. However, to sustain proper neural activity in the brain, another, no less important network is operating: the vast, complex and fascinating vascular network. Increasing research has characterized schizophrenia as a systemic disease where vascular involvement is important. Several neuro-angiogenic pathway disturbances have been related to schizophrenia. Alterations, ranging from genetic polymorphisms, mRNA, and protein alterations to microRNA and abnormal metabolite processing, have been evaluated in plasma, post-mortem brain, animal models, and patient-derived induced pluripotent stem cell (hiPSC) models. During embryonic brain development, the coordinated formation of blood vessels parallels neuro/gliogenesis and results in the structuration of the neurovascular niche, which brings together physical and molecular signals from both systems conforming to the Blood-Brain barrier. In this review, we offer an upfront perspective on distinctive angiogenic and neurogenic signaling pathways that might be involved in the biological causality of schizophrenia. We analyze the role of pivotal angiogenic-related pathways such as Vascular Endothelial Growth Factor and HIF signaling related to hypoxia and oxidative stress events; classic developmental pathways such as the NOTCH pathway, metabolic pathways such as the mTOR/AKT cascade; emerging neuroinflammation, and neurodegenerative processes such as UPR, and also discuss non-canonic angiogenic/axonal guidance factor signaling. Considering that all of the mentioned above pathways converge at the Blood-Brain barrier, reported neurovascular alterations could have deleterious repercussions on overall brain functioning in schizophrenia.
Collapse
|
23
|
Lasa M, Contreras-Jurado C. Thyroid hormones act as modulators of inflammation through their nuclear receptors. Front Endocrinol (Lausanne) 2022; 13:937099. [PMID: 36004343 PMCID: PMC9393327 DOI: 10.3389/fendo.2022.937099] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Reciprocal crosstalk between endocrine and immune systems has been well-documented both in physiological and pathological conditions, although the connection between the immune system and thyroid hormones (THs) remains largely unclear. Inflammation and infection are two important processes modulated by the immune system, which have profound effects on both central and peripheral THs metabolism. Conversely, optimal levels of THs are necessary for the maintenance of immune function and response. Although some effects of THs are mediated by their binding to cell membrane integrin receptors, triggering a non-genomic response, most of the actions of these hormones involve their binding to specific nuclear thyroid receptors (TRs), which generate a genomic response by modulating the activity of a great variety of transcription factors. In this special review on THs role in health and disease, we highlight the relevance of these hormones in the molecular mechanisms linked to inflammation upon their binding to specific nuclear receptors. In particular, we focus on THs effects on different signaling pathways involved in the inflammation associated with various infectious and/or pathological processes, emphasizing those mediated by NF-kB, p38MAPK and JAK/STAT. The findings showed in this review suggest new opportunities to improve current therapeutic strategies for the treatment of inflammation associated with several infections and/or diseases, such as cancer, sepsis or Covid-19 infection.
Collapse
Affiliation(s)
- Marina Lasa
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Constanza Contreras-Jurado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Alfonso X El Sabio, Madrid, Spain
- Departamento de Fisiopatología Endocrina y del Sistema Nervioso, Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
24
|
Examination of Combined Treatment of Ginsenoside Rg3 and 5-Fluorouracil in Lung Adenocarcinoma Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2813142. [PMID: 35799655 PMCID: PMC9256322 DOI: 10.1155/2022/2813142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/15/2022] [Indexed: 12/21/2022]
Abstract
Chemotherapy is a commonly used strategy for advanced lung cancer patients. However, its clinical application is restrained due to its toxicity and drug resistance. Ginsenoside Rg3 (Rg3) has a strong anticancer influence on colon cancer, breast cancer, lung cancer, and other malignant tumors. However, it is still unclear whether Rg3 can cooperate with 5-FU to inhibit the tumor growth and angiogenesis of lung adenocarcinoma (LUAD). This study examined the combined treatment of Rg3 and 5-FU in LUAD. It was revealed that the combined treatment could notably enhance the suppression on proliferative, invasive, and migratory abilities and angiogenesis in LUAD cells A549 and SPC-A-1. On the other hand, we also discovered that Rg3 or 5-FU could suppress the activity of the NF-κB signaling pathway and downregulate VEGFA expression in LUAD cells. Collectively, this study suggested that Rg3 combined chemotherapy may perform a more powerful drug efficiency in LUAD cells.
Collapse
|
25
|
Bakshi HA, Quinn GA, Nasef MM, Mishra V, Aljabali AAA, El-Tanani M, Serrano-Aroca Á, Webba Da Silva M, McCarron PA, Tambuwala MM. Crocin Inhibits Angiogenesis and Metastasis in Colon Cancer via TNF-α/NF-kB/VEGF Pathways. Cells 2022; 11:1502. [PMID: 35563808 PMCID: PMC9104358 DOI: 10.3390/cells11091502] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and metastasis play pivotal roles in the progression of cancer. We recently discovered that crocin, a dietary carotenoid derived from the Himalayan crocus, inhibited the growth of colon cancer cells. However, the exact role of crocin on the angiogenesis and metastasis in colorectal cancer remains unclear. In the present study, we demonstrated that crocin significantly reduces the viability of colon cancer cells (HT-29, Caco-2) and human umbilical vein endothelial cells (HUVEC), but was not toxic to human colon epithelial (HCEC) cells. Furthermore, pre-treatment of human carcinoma cells (HT-29 and Caco-2) with crocin inhibited cell migration, invasion, and angiogenesis in concentration -dependent manner. Further studies demonstrated that crocin inhibited TNF-α, NF-κB and VEGF pathways in colon carcinoma cell angiogenesis and metastasis. Crocin also inhibited cell migration, invasion, and tube formation in human umbilical vein endothelial cells (HUVEC) in a concentration -dependent manner. We also observed that crocin significantly reduced the secretion of VEGF and TNF-α induced activation of NF-kB by human colon carcinoma cells. In the absence of TNF-α, a concentration-dependent reduction in NF-kB was observed. Many of these observations were confirmed by in vivo angiogenesis models, which showed that crocin significantly reduced the progression of tumour growth. Collectively, these finding suggest that crocin inhibits angiogenesis and colorectal cancer cell metastasis by targeting NF-kB and blocking TNF-α/NF-κB/VEGF pathways.
Collapse
Affiliation(s)
- Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| | - Gerry A. Quinn
- School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
| | - Mohamed M. Nasef
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 566, Jordan;
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Mateus Webba Da Silva
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| | - Paul A. McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| |
Collapse
|
26
|
De S, Tamagno I, Stark GR, Jackson MW. Validation-Based Insertional Mutagenesis (VBIM), A Powerful Forward Genetic Screening Strategy. Curr Protoc 2022; 2:e394. [PMID: 35316583 PMCID: PMC8969887 DOI: 10.1002/cpz1.394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Forward genetics begins with a biological phenotype and attempts to identify genetic changes that influence that phenotype. These changes can be induced in a selected group of genes, for instance, by using libraries of cDNAs, shRNAs, CRISPR guide RNAs, or genetic suppressor elements (GSEs), or randomly throughout the genome using chemical or insertional mutagens, with each approach creating distinct genetic changes. The Validation-Based Insertional Mutagenesis (VBIM) strategy utilizes modified lentiviruses as insertional mutagens, placing strong promoters throughout the genome. Generating libraries with millions of cells carrying one or a few VBIM promoter insertions is straightforward, allowing selection of cells in which overexpression of VBIM-driven RNAs or proteins promote the phenotype of interest. VBIM-driven RNAs may encode full-length proteins, truncated proteins (which may have wild-type, constitutive, or dominant-negative activity), or antisense RNAs that can disrupt gene expression. The diversity in VBIM-driven changes allows for the identification of both gain-of-function and loss-of-function mutations in a single screen. Additionally, VBIM can target any genomic locus, regardless of whether it is expressed in the cells under study or known to have a biological function, allowing for true whole-genome screens without the complication and cost of constructing, maintaining, and delivering a comprehensive library. Here, we review the VBIM strategy and discuss examples in which VBIM has been successfully used in diverse screens to identify novel genes or novel functions for known genes. In addition, we discuss considerations for transitioning the VBIM strategy to in vivo screens. We hope that other laboratories will be encouraged to use the VBIM strategy to identify genes that influence their phenotypes of interest. © 2022 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Sarmishtha De
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - George R Stark
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106,Corresponding author: Mark W. Jackson, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106. Tel: 216.368.1276,
| |
Collapse
|
27
|
Niknam Z, Samadi M, Ghalibafsabbaghi A, Chodari L. IGF-I combined with exercise improve diabetes-induced vascular dysfunction in heart of male Wistar rats. J Cardiovasc Thorac Res 2021; 14:34-41. [PMID: 35620752 PMCID: PMC9106942 DOI: 10.34172/jcvtr.2021.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction: This research investigates the impact of insulin-like growth factor-I (IGF -I)and exercise on mediators associated with angiogenesis (VEGF-A, TSP-1, and NF-кβ) and capillarization status of the diabetic rats’ hearts.
Methods: Splitting of forty Wistar male rats into five groups occurred as following: control,diabetes, diabetes+IGF-I, diabetes+exercise, and diabetes+exercise+IGF-I.Through intraperitoneal administration of 60 mg/kg streptozotocin, the condition of Type 1diabetes was escalated. After four weeks of treatment with IGF-I (2 mg/kg/day) or treadmill exercise (17 m/min, zero degrees slope, 30 min/day), in the heart, microvascular density and protein levels of VEGF-A, TSP-1, and NF-кβ were determined by H&E staining and ELISA,respectively.
Results: Within the diabetic group, observations present a significant decrease in VEGF-A and MVD levels, whereas an increase in the TSP-1 and NF-Κb levels. While these impacts were reversed by either IGF-I or exercise treatments, simultaneous treatment had synergistic effects. Moreover, among diabetic rats, undesirable histologic alterations of the heart were demonstrated, including myonecrosis, interstitial edema, hemorrhage, and mononuclear immune cell infiltration, whereas treatments improved these changes.
Conclusion: These data manifest that IGF-I and exercise can increase the cardiac angiogenesis of diabetic rats through increasing expression of VEGF-A, and decreasing TSP-1 and NF-кβproteins level, also can improve myocardial tissue damages.
Collapse
Affiliation(s)
- Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahrokh Samadi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences , Urmia, Iran
| | | | - leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
28
|
Silke J, O’Reilly LA. NF-κB and Pancreatic Cancer; Chapter and Verse. Cancers (Basel) 2021; 13:4510. [PMID: 34572737 PMCID: PMC8469693 DOI: 10.3390/cancers13184510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the world's most lethal cancers. An increase in occurrence, coupled with, presently limited treatment options, necessitates the pursuit of new therapeutic approaches. Many human cancers, including PDAC are initiated by unresolved inflammation. The transcription factor NF-κB coordinates many signals that drive cellular activation and proliferation during immunity but also those involved in inflammation and autophagy which may instigate tumorigenesis. It is not surprising therefore, that activation of canonical and non-canonical NF-κB pathways is increasingly recognized as an important driver of pancreatic injury, progression to tumorigenesis and drug resistance. Paradoxically, NF-κB dysregulation has also been shown to inhibit pancreatic inflammation and pancreatic cancer, depending on the context. A pro-oncogenic or pro-suppressive role for individual components of the NF-κB pathway appears to be cell type, microenvironment and even stage dependent. This review provides an outline of NF-κB signaling, focusing on the role of the various NF-κB family members in the evolving inflammatory PDAC microenvironment. Finally, we discuss pharmacological control of NF-κB to curb inflammation, focussing on novel anti-cancer agents which reinstate the process of cancer cell death, the Smac mimetics and their pre-clinical and early clinical trials.
Collapse
Affiliation(s)
- John Silke
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Lorraine Ann O’Reilly
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
29
|
Chemotherapy: a double-edged sword in cancer treatment. Cancer Immunol Immunother 2021; 71:507-526. [PMID: 34355266 DOI: 10.1007/s00262-021-03013-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Chemotherapy is a well-known and effective treatment for different cancers; unfortunately, it has not been as efficient in the eradication of all cancer cells as been expected. The mechanism of this failure was not fully clarified, yet. Meanwhile, alterations in the physiologic conditions of the tumor microenvironment (TME) were suggested as one of the underlying possibilities. Chemotherapy drugs can activate multiple signaling pathways and augment the secretion of inflammatory mediators. Inflammation may show two opposite roles in the TME. On the one hand, inflammation, as an innate immune response, tries to suppress tumor growth but on the other hand, it might be not powerful enough to eradicate the cancer cells and even it can provide appropriate conditions for cancer promotion and relapse as well. Therefore, the administration of mild anti-inflammatory drugs during chemotherapy might result in more successful clinical results. Here, we will review and discuss this hypothesis. Most chemotherapy agents are triggers of inflammation in the tumor microenvironment through inducing the production of senescence-associated secretory phenotype (SASP) molecules. Some chemotherapy agents can induce systematic inflammation by provoking TLR4 signaling or triggering IL-1B secretion through the inflammasome pathway. NF-kB and MAPK are key signaling pathways of inflammation and could be activated by several chemotherapy drugs. Furthermore, inflammation can play a key role in cancer development, metastasis and exacerbation.
Collapse
|
30
|
Gilmore TD. NF-κB and Human Cancer: What Have We Learned over the Past 35 Years? Biomedicines 2021; 9:biomedicines9080889. [PMID: 34440093 PMCID: PMC8389606 DOI: 10.3390/biomedicines9080889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
Transcription factor NF-κB has been extensively studied for its varied roles in cancer development since its initial characterization as a potent retroviral oncogene. It is now clear that NF-κB also plays a major role in a large variety of human cancers, including especially ones of immune cell origin. NF-κB is generally constitutively or aberrantly activated in human cancers where it is involved. These activations can occur due to mutations in the NF-κB transcription factors themselves, in upstream regulators of NF-κB, or in pathways that impact NF-κB. In addition, NF-κB can be activated by tumor-assisting processes such as inflammation, stromal effects, and genetic or epigenetic changes in chromatin. Aberrant NF-κB activity can affect many tumor-associated processes, including cell survival, cell cycle progression, inflammation, metastasis, angiogenesis, and regulatory T cell function. As such, inhibition of NF-κB has often been investigated as an anticancer strategy. Nevertheless, with a few exceptions, NF-κB inhibition has had limited success in human cancer treatment. This review covers general themes that have emerged regarding the biological roles and mechanisms by which NF-κB contributes to human cancers and new thoughts on how NF-κB may be targeted for cancer prognosis or therapy.
Collapse
|
31
|
Darbuka E, Gürkaşlar C, Yaman I. Ochratoxin A induces ERK1/2 phosphorylation-dependent apoptosis through NF-κB/ERK axis in human proximal tubule HK-2 cell line. Toxicon 2021; 199:79-86. [PMID: 34116085 DOI: 10.1016/j.toxicon.2021.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/15/2021] [Accepted: 06/04/2021] [Indexed: 12/08/2022]
Abstract
Ochratoxin A (OTA) is a food contaminant mycotoxin with hazardous effects on human and animal health, primarily affecting the kidneys. OTA's mode of action is not well understood. OTA activates both MAPK/ERK and PI3K/Akt signaling pathways, which play role in apoptosis and cell survival, respectively. OTA is also known to induce toxicity by activating the NF-κB pathway in immune cells. However, its role in determining the cell fate upon OTA exposure in a human kidney cell line (HK-2) has not been fully explored. We made use of pharmacological inhibition of NF-κB to define its role in viability of OTA-treated HK-2 cells. We show that OTA-induced p65 NF-κB subunit translocation into the nucleus in a time-dependent manner using both Western blotting and immunofluorescence (IF). We also document the DNA-binding and reporter gene expression activities of NF-κB by electrophoretic mobility shift (EMSA) and luciferase reporter assays, respectively. Our results indicate that, following 6 h of exposure, OTA fully activates NF-κB pathway and its downstream effectors in HK-2 cells. In addition, Bay11-7085 treatment causes attenuation of the relative levels of OTA-mediated ERK1/2 phosphorylation, suggesting a cross-talk between NF-κB and the MAPK/ERK pathway. Critically, co-treatment of HK-2 cells with OTA and Bay11-7085 leads to the inhibition of OTA-induced apoptosis in a time-dependent manner. Our results support a robust association between NF-κB and the MAPK/ERK pathways in the modulation of apoptotic effects of OTA in HK-2 cells.
Collapse
Affiliation(s)
- Elif Darbuka
- Bogazici University, Department of Molecular Biology and Genetics, Molecular Toxicology and Cancer Research Laboratory, Bebek, Istanbul, 34342, Turkey
| | - Can Gürkaşlar
- Bogazici University, Department of Molecular Biology and Genetics, Molecular Toxicology and Cancer Research Laboratory, Bebek, Istanbul, 34342, Turkey
| | - Ibrahim Yaman
- Bogazici University, Department of Molecular Biology and Genetics, Molecular Toxicology and Cancer Research Laboratory, Bebek, Istanbul, 34342, Turkey; Bogazici University, Center for Life Sciences and Technologies, Bebek, Istanbul, 34342, Turkey.
| |
Collapse
|
32
|
Wijaya LS, Trairatphisan P, Gabor A, Niemeijer M, Keet J, Alcalà Morera A, Snijders KE, Wink S, Yang H, Schildknecht S, Stevens JL, Bouwman P, Kamp H, Hengstler J, Beltman J, Leist M, Le Dévédec S, Saez-Rodriguez J, van de Water B. Integration of temporal single cell cellular stress response activity with logic-ODE modeling reveals activation of ATF4-CHOP axis as a critical predictor of drug-induced liver injury. Biochem Pharmacol 2021; 190:114591. [PMID: 33957093 DOI: 10.1016/j.bcp.2021.114591] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022]
Abstract
Drug-induced liver injury (DILI) is the most prevalent adversity encountered in drug development and clinical settings leading to urgent needs to understand the underlying mechanisms. In this study, we have systematically investigated the dynamics of the activation of cellular stress response pathways and cell death outcomes upon exposure of a panel of liver toxicants using live cell imaging of fluorescent reporter cell lines. We established a comprehensive temporal dynamic response profile of a large set of BAC-GFP HepG2 cell lines representing the following components of stress signaling: i) unfolded protein response (UPR) [ATF4, XBP1, BIP and CHOP]; ii) oxidative stress [NRF2, SRXN1, HMOX1]; iii) DNA damage [P53, P21, BTG2, MDM2]; and iv) NF-κB pathway [A20, ICAM1]. We quantified the single cell GFP expression as a surrogate for endogenous protein expression using live cell imaging over > 60 h upon exposure to 14 DILI compounds at multiple concentrations. Using logic-based ordinary differential equation (Logic-ODE), we modelled the dynamic profiles of the different stress responses and extracted specific descriptors potentially predicting the progressive outcomes. We identified the activation of ATF4-CHOP axis of the UPR as the key pathway showing the highest correlation with cell death upon DILI compound perturbation. Knocking down main components of the UPR provided partial protection from compound-induced cytotoxicity, indicating a complex interplay among UPR components as well as other stress pathways. Our results suggest that a systematic analysis of the temporal dynamics of ATF4-CHOP axis activation can support the identification of DILI risk for new candidate drugs.
Collapse
Affiliation(s)
- Lukas Surya Wijaya
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Panuwat Trairatphisan
- Heidelberg University, Faculty of Medicine, Institute of Computational Biomedicine, 69120 Heidelberg, Germany
| | - Attila Gabor
- Heidelberg University, Faculty of Medicine, Institute of Computational Biomedicine, 69120 Heidelberg, Germany
| | - Marije Niemeijer
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Jason Keet
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Ariadna Alcalà Morera
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Kirsten E Snijders
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Steven Wink
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Huan Yang
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Stefan Schildknecht
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - James L Stevens
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Peter Bouwman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Hennicke Kamp
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen am Rhein, Germany
| | - Jan Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Joost Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Sylvia Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, Institute of Computational Biomedicine, 69120 Heidelberg, Germany; RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), 52074 Aachen, Germany
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
33
|
Fordjour FA, Asiedu E, Larbi A, Kwarteng A. The role of nuclear factor kappa B (NF-κB) in filarial pathology. J Cell Commun Signal 2021; 15:185-193. [PMID: 33630268 DOI: 10.1007/s12079-021-00607-5] [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: 12/28/2020] [Accepted: 01/24/2021] [Indexed: 12/11/2022] Open
Abstract
The transcription factor NF-κB promotes immunity by controlling the expression of genes involved in inflammation. Cytokines and pathogen-associated molecular patterns stimulate cell surface receptors, including toll-like receptors, to initiate a signalling cascade resulting in the activation of NF-κB. NF-κB drives the expression of target genes that mediate cell proliferation and release antimicrobial molecules and cytokines to activate an immune response. Filariasis is one of the most complex infections of humans. The actual causes of the heterogeneity in infection are not well understood. However, they have been attributed to differences in inflammatory processes that are immune-mediated, secondary bacterial infections, and host immune-genetics. Elevated production of angiogenic molecules (VEGFs, CEACAM and MMPs) in filarial pathology has been shown to be dependent on phosphorylation and intracellular activation of NF-κB. This review examines the role of NF-κB in filarial pathology and its potential therapeutic options for individuals with the disease.
Collapse
Affiliation(s)
- Fatima Amponsah Fordjour
- Department of Microbiology, University for Development Studies, UDS, Tamale, Ghana. .,Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, KNUST, Kumasi, Ghana.
| | - Ebenezer Asiedu
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, KNUST, Kumasi, Ghana
| | - Amma Larbi
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, KNUST, Kumasi, Ghana
| | - Alexander Kwarteng
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, KNUST, Kumasi, Ghana.,Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, KNUST, Kumasi, Ghana
| |
Collapse
|
34
|
Jiang Y, Müller K, Khan MA, Assmann JC, Lampe J, Kilau K, Richter M, Kleint M, Ridder DA, Hübner N, Schmidt-Supprian M, Wenzel J, Schwaninger M. Cerebral angiogenesis ameliorates pathological disorders in Nemo-deficient mice with small-vessel disease. J Cereb Blood Flow Metab 2021; 41:219-235. [PMID: 32151223 PMCID: PMC8369998 DOI: 10.1177/0271678x20910522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cerebral small-vessel diseases (SVDs) often follow a progressive course. Little is known about the function of angiogenesis, which potentially induces regression of SVDs. Here, we investigated angiogenesis in a mouse model of incontinentia pigmenti (IP), a genetic disease comprising features of SVD. IP is caused by inactivating mutations of Nemo, the essential component of NF-κB signaling. When deleting Nemo in the majority of brain endothelial cells (NemobeKO mice), the transcriptional profile of vessels indicated cell proliferation. Brain endothelial cells expressed Ki67 and showed signs of DNA synthesis. In addition to cell proliferation, we observed sprouting and intussusceptive angiogenesis in NemobeKO mice. Angiogenesis occurred in all segments of the vasculature and in proximity to vessel rarefaction and tissue hypoxia. Apparently, NEMO was required for productive angiogenesis because endothelial cells that had escaped Nemo inactivation showed a higher proliferation rate than Nemo-deficient cells. Therefore, newborn endothelial cells were particularly vulnerable to ongoing recombination. When we interfered with productive angiogenesis by inducing ongoing ablation of Nemo, mice did not recover from IP manifestations but rather showed severe functional deficits. In summary, the data demonstrate that angiogenesis is present in this model of SVD and suggest that it may counterbalance the loss of vessels.
Collapse
Affiliation(s)
- Yun Jiang
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Kristin Müller
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Mahtab A Khan
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Julian C Assmann
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Josephine Lampe
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Germany
| | - Knut Kilau
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Germany
| | - Marius Richter
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Maximilian Kleint
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Dirk A Ridder
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Norbert Hübner
- DZHK (German Research Centre for Cardiovascular Research), Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Germany
| |
Collapse
|
35
|
Yee YH, Chong SJF, Kong LR, Goh BC, Pervaiz S. Sustained IKKβ phosphorylation and NF-κB activation by superoxide-induced peroxynitrite-mediated nitrotyrosine modification of B56γ3 and PP2A inactivation. Redox Biol 2020; 41:101834. [PMID: 33838472 PMCID: PMC8056462 DOI: 10.1016/j.redox.2020.101834] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 01/15/2023] Open
Abstract
Apart from its physiological role in inflammation and immunity, the nuclear factor-kappa B (NF-κB) protein complex has been implicated in tumorigenesis and its progression. Here, we provide evidence that a pro-oxidant milieu is an upstream effector of oncogenic NF-κB signaling. Through pharmacological or genetic inhibition of SOD1, we show that elevated intracellular superoxide (O2•-) mediates sustained IKK phosphorylation, and induces downstream degradation of IκBα, leading to the nuclear localization and transcriptional activation of NF-κB. Mechanistically, we show that such sustained NF-κB signaling is a function of protein phosphatase 2A (PP2A) inactivation brought about by the nitrative modification of its substrate-binding sub-unit B56γ. Importantly, the pro-oxidant driven NF-κB activation enhances the migratory and invasive potential of cancer cells. In summary, our work highlights the critical involvement of O2•--dependent peroxynitrite production in inhibiting PP2A-mediated dephosphorylation of IKK, thereby facilitating cancers to acquire an invasive phenotype. Given that NF-κB is a key player of chronic inflammation and carcinogenesis, our work unravels a novel synergistic node involving O2•--driven redox milieu and deregulated PP2A as a potential therapeutic target.
Collapse
Affiliation(s)
- Yi Hui Yee
- Cancer Science Institute of Singapore, National University of Singapore (NUS), Singapore
| | | | - Li Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore (NUS), Singapore; Medical Research Council Cancer Unit, University of Cambridge, Cambridge, CB2, 0XZ, United Kingdom
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore (NUS), Singapore; Department of Hematology-Oncology, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, NUS, Singapore; National University Cancer Institute, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Shazib Pervaiz
- National University Cancer Institute, National University Health System, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore; Integrative Science and Engineering Programme, NUS Graduate School, NUS, Singapore; Faculté de Medicine, University of Paris, Paris, France.
| |
Collapse
|
36
|
Teppan J, Barth DA, Prinz F, Jonas K, Pichler M, Klec C. Involvement of Long Non-Coding RNAs (lncRNAs) in Tumor Angiogenesis. Noncoding RNA 2020; 6:E42. [PMID: 32992718 PMCID: PMC7711482 DOI: 10.3390/ncrna6040042] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are defined as non-protein coding transcripts with a minimal length of 200 nucleotides. They are involved in various biological processes such as cell differentiation, apoptosis, as well as in pathophysiological processes. Numerous studies considered that frequently deregulated lncRNAs contribute to all hallmarks of cancer including metastasis, drug resistance, and angiogenesis. Angiogenesis, the formation of new blood vessels, is crucial for a tumor to receive sufficient amounts of nutrients and oxygen and therefore, to grow and exceed in its size over the diameter of 2 mm. In this review, the regulatory mechanisms of lncRNAs are described, which influence tumor angiogenesis by directly or indirectly regulating oncogenic pathways, interacting with other transcripts such as microRNAs (miRNAs) or modulating the tumor microenvironment. Further, angiogenic lncRNAs occurring in several cancer types such as liver, gastrointestinal cancer, or brain tumors are summarized. Growing evidence on the influence of lncRNAs on tumor angiogenesis verified these transcripts as potential predictive or diagnostic biomarkers or therapeutic targets of anti-angiogenesis treatment. However, there are many unsolved questions left which are pointed out in this review, hence driving comprehensive research in this area is necessary to enable an effective use of lncRNAs as either therapeutic molecules or diagnostic targets in cancer.
Collapse
Affiliation(s)
- Julia Teppan
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
| | - Dominik A. Barth
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Felix Prinz
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
| | - Katharina Jonas
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christiane Klec
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; (J.T.); (D.A.B.); (F.P.); (K.J.); (C.K.)
| |
Collapse
|
37
|
Zaw SYM, Kaneko T, Zaw ZCT, Sone PP, Murano H, Gu B, Okada Y, Han P, Katsube KI, Okiji T. Crosstalk between dental pulp stem cells and endothelial cells augments angiogenic factor expression. Oral Dis 2020; 26:1275-1283. [PMID: 32248596 DOI: 10.1111/odi.13341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES We aimed to investigate whether the mesenchymal stem cell-endothelial cell crosstalk enhances angiogenic factor expression via nuclear factor-kappa B (NF-κB)-dependent mechanisms. MATERIALS AND METHODS Human dermal microvascular endothelial cells (HDMECs) and stem cells from human exfoliated deciduous teeth (SHEDs) were cocultured for 96 hr, in the presence of NF-κB decoy oligodeoxynucleotides (ODNs) or scramble (control). Vascular endothelial cell growth factor (VEGF) and phospho-NF-κB p65 were measured with enzyme-linked immunosorbent assay. Angiogenesis-related gene expression was analyzed with microarray analysis followed by real-time polymerase chain reaction. Tube formation assay was conducted in the presence of NF-κB decoy. RESULTS The VEGF and phospho-NF-κB p65 levels were significantly higher in the coculture with NF-κB decoy scramble than in single culture and coculture with NF-κB decoy ODN. Microarray analysis of SHEDs and HDMECs with NF-κB decoy scramble showed higher expression of proangiogenic genes, Bcl-2, NF-κB1, VEGFA, CXCL8, and CXCR1, and lower expression of proapoptotic genes, Bax and Caspase 9, compared to cells with NF-κB decoy ODN. Real-time PCR results for Bcl-2 and CXCL8 showed a similar trend. Tube formation assay showed more tube development in the presence of NF-κB decoy scramble. CONCLUSION The SHED-HDMEC crosstalk enhanced proangiogenic factor expression via NF-κB-dependent pathways.
Collapse
Affiliation(s)
- Su Yee Myo Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomoatsu Kaneko
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Zar Chi Thein Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Phyo Pyai Sone
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroki Murano
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Bin Gu
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yamato Okada
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Peifeng Han
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
38
|
NF-κB inhibitors in treatment and prevention of lung cancer. Biomed Pharmacother 2020; 130:110569. [PMID: 32750649 DOI: 10.1016/j.biopha.2020.110569] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/12/2020] [Accepted: 07/26/2020] [Indexed: 12/27/2022] Open
Abstract
Intracellular signalling pathways have provided excellent resource for drug development particularly in the development of cancer therapeutics. A wide variety of malignancies common in human exhibit aberrant NF-κB constitutive expression which results in tumorigenic processes and cancer survival in a variety of solid tumour, including pancreatic cancer, lung, cervical, prostate, breast and gastric carcinoma. Numerous evidences indicate that NF-κB signalling mechanism is mainly involved in the progression of several cancers which may intensify an enhanced knowledge on its role in disease particularly lung tumorigenesis. This has led to tremendous research in designing a variety of NF-κB antagonists with enhanced clinical applications through different approaches the most common being suppression of IκB kinase (IKK) beta activity. Many NF-κB inhibitors for lung cancer are now under clinical trials. Preliminary results of clinical trials for several of these agents include small-molecule inhibitors and monoclonal antibodies. A few combinatorial treatment therapies are currently under investigation in the clinics and have shown promise, particularly NF-κB inhibition associated with lung cancer.
Collapse
|
39
|
Apoptotic exosome-like vesicles regulate endothelial gene expression, inflammatory signaling, and function through the NF-κB signaling pathway. Sci Rep 2020; 10:12562. [PMID: 32724121 PMCID: PMC7387353 DOI: 10.1038/s41598-020-69548-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Persistent endothelial injury promotes maladaptive responses by favoring the release of factors leading to perturbation in vascular homeostasis and tissue architecture. Caspase-3 dependent death of microvascular endothelial cells leads to the release of unique apoptotic exosome-like vesicles (ApoExo). Here, we evaluate the impact of ApoExo on endothelial gene expression and function in the context of a pro-apoptotic stimulus. Endothelial cells exposed to ApoExo differentially express genes involved in cell death, inflammation, differentiation, and cell movement. Endothelial cells exposed to ApoExo showed inhibition of apoptosis, improved wound closure along with reduced angiogenic activity and reduced expression of endothelial markers consistent with the first phase of endothelial-to-mesenchymal transition (endoMT). ApoExo interaction with endothelial cells also led to NF-κB activation. NF-κB is known to participate in endothelial dysfunction in numerous diseases. Silencing NF-κB reversed the anti-apoptotic effect and the pro-migratory state and prevented angiostatic properties and CD31 downregulation in endothelial cells exposed to ApoExo. This study identifies vascular injury-derived extracellular vesicles (ApoExo) as novel drivers of NF-κB activation in endothelial cells and demonstrates the pivotal role of this signaling pathway in coordinating ApoExo-induced functional changes in endothelial cells. Hence, targeting ApoExo-mediated NF-κB activation in endothelial cells opens new avenues to prevent endothelial dysfunction.
Collapse
|
40
|
Harmsen MJ, Wong CFC, Mijatovic V, Griffioen AW, Groenman F, Hehenkamp WJK, Huirne JAF. Role of angiogenesis in adenomyosis-associated abnormal uterine bleeding and subfertility: a systematic review. Hum Reprod Update 2020; 25:647-671. [PMID: 31504506 PMCID: PMC6737562 DOI: 10.1093/humupd/dmz024] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Adenomyosis commonly occurs with abnormal uterine bleeding (AUB) and is associated with subfertility and a higher miscarriage rate. Recent evidence showed abnormal vascularization in the endometrium in patients with adenomyosis, suggesting a role of angiogenesis in the pathophysiology of AUB and subfertility in adenomyosis and providing a possible treatment target. OBJECTIVE AND RATIONALE We hypothesized that the level of abnormal vascularization and expression of angiogenic markers is increased in the ectopic and eutopic endometrium of adenomyosis patients in comparison with the endometrium of control patients. This was investigated through a search of the literature. SEARCH METHODS A systematic search was performed in PubMed and Embase until February 2019. Combinations of terms for angiogenesis and adenomyosis were applied as well as AUB, subfertility or anti-angiogenic therapy. The main search was limited to clinical studies carried out on premenopausal women. Original research articles focusing on markers of angiogenesis in the endometrium of patients with adenomyosis were included. Studies in which no comparison was made to control patients or which were not published in a peer-reviewed journal were excluded. A second search was performed to explore the therapeutic potential of targeting angiogenesis in adenomyosis. This search also included preclinical studies. OUTCOMES A total of 20 articles out of 1669 hits met our selection criteria. The mean vascular density (MVD) was studied by quantification of CD31, CD34, von Willebrand Factor (vWF) or factor-VIII-antibody-stained microvessels in seven studies. All these studies reported a significantly increased MVD in ectopic endometrium, and out of the six articles that took it into account, four studies reported a significantly increased MVD in eutopic endometrium compared with control endometrium. Five articles showed a significantly higher vascular endothelial growth factor expression in ectopic endometrium and three articles in eutopic endometrium compared with control endometrium. The vascular and pro-angiogenic markers α-smooth muscle actin, endoglin, S100A13, vimentin, matrix metalloproteinases (MMPs), nuclear factor (NF)-kB, tissue factor (TF), DJ-1, phosphorylated mammalian target of rapamycin, activin A, folli- and myostatin, CD41, SLIT, roundabout 1 (ROBO1), cyclooxygenase-2, lysophosphatidic acid (LPA) 1,4-5, phospho signal transducer and activator of transcription 3 (pSTAT3), interleukin (IL)-6, IL-22 and transforming growth factor-β1 were increased in ectopic endometrium, and the markers S100A13, MMP-2 and -9, TF, follistatin, myostatin, ROBO1, LPA1 and 4-5, pSTAT3, IL-6 and IL-22 were increased in eutopic endometrium, compared with control endometrium. The anti-angiogenic markers E-cadherin, eukaryotic translation initiation factor 3 subunit and gene associated with retinoic-interferon-induced mortality 19 were decreased in ectopic endometrium and IL-10 in eutopic endometrium, compared with control endometrium. The staining level of vWF and two pro-angiogenic markers (NF-κB nuclear p65 and TF) correlated with AUB in patients with adenomyosis. We found no studies that investigated the possible relationship between markers of angiogenesis and subfertility in adenomyosis patients. Nine articles reported on direct or indirect targeting of angiogenesis in adenomyosis-either by testing hormonal therapy or herbal compounds in clinical studies or by testing angiogenesis inhibitors in preclinical studies. However, there are no clinical studies on the effectiveness of such therapy for adenomyosis-related AUB or subfertility. WIDER IMPLICATIONS The results are in agreement with our hypothesis that increased angiogenesis is present in the endometrium of patients with adenomyosis compared with the endometrium of control patients. It is likely that increased angiogenesis leads to fragile and more permeable vessels resulting in adenomyosis-related AUB and possibly subfertility. While this association has not sufficiently been studied yet, our results encourage future studies to investigate the exact role of angiogenesis in the etiology of adenomyosis and related AUB or subfertility in women with adenomyosis in order to design curative or preventive therapeutic strategies.
Collapse
Affiliation(s)
- Marissa J Harmsen
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Caroline F C Wong
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Velja Mijatovic
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Freek Groenman
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Wouter J K Hehenkamp
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Pourheydar B, Biabanghard A, Azari R, Khalaji N, Chodari L. Exercise improves aging-related decreased angiogenesis through modulating VEGF-A, TSP-1 and p-NF-Ƙb protein levels in myocardiocytes. J Cardiovasc Thorac Res 2020; 12:129-135. [PMID: 32626553 PMCID: PMC7321007 DOI: 10.34172/jcvtr.2020.21] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction: Aging-dependent decline in the angiogenesis of heart is a risk factor for cardiovascular disease. This study was aimed to characterize effect of exercise on angiogenesis alterations and molecular mediators which are related to angiogenesis in the heart under aging condition. Methods: Twenty-one male Wistar rats were assigned into three groups: young, aged, and exercise. Aged animals in the exercise group run on treadmill for 8 weeks. At the end, heart samples were collected and used for histological evaluation , determination of angiogenesis by immunostaining for PECAM-1/ CD31 and expressions of vascular endothelial growth factor-A (VEGF-A), thrombospondin-1 (TSP-1) and nuclear factor kappa B (NF-κB) levels by ELISA. P<0.05 is considered as statistically significant. Results: Our results showed that angiogenesis, and VEGF-A levels were significantly decreased, TSP1 (P >0.0001) and p-NF-κB (P >0.001) levels were significantly increased in the heart of aged group compared to young group. Exercise group showed significant increase in angiogenesis, VEGF-A (P >0.0001), and p-NF-κB (P >0.001) and showed significant decrease in TSP-1 levels (P >0.001) compared to aged group. Moreover, compared to the young group, aged group showed histological changes in the heart, such as interstitial edema, and congestion, whereas, treatment with exercise improved these undesirable changes in the heart of exercise groups. Conclusion: These findings indicated that aging-related decrease in angiogenesis in the heart may mediated by downexpression of VEGF-A and overexpression of TSP-1 proteins. Also, we showed that p-NF-κB protein was increased in the heart of aged rats, this probably mediated by compensatory mechanism. It was also showed that exercise as novel non-pharmacological therapy modifies VEGF-A and TSP-1 and increases p-NF-κB protein levels in the aged heart.
Collapse
Affiliation(s)
- Bagher Pourheydar
- Department of Anatomical Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute,Urmia University of Medical Sciences, Urmia, Iran
| | - Abdolrahman Biabanghard
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Azari
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Naser Khalaji
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute,Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
42
|
Saboory E, Gholizadeh-Ghaleh Aziz S, Samadi M, Biabanghard A, Chodari L. Exercise and insulin-like growth factor 1 supplementation improve angiogenesis and angiogenic cytokines in a rat model of diabetes-induced neuropathy. Exp Physiol 2020; 105:783-792. [PMID: 32053260 DOI: 10.1113/ep088069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/11/2020] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? Do changes in levels of angiogenesis-related mediators [vascular endothelial growth factor-A (VEGF-A), thrombospondin-1 (TSP-1) and nuclear factor-κB (NF-κB)] in the sciatic nerve mediate diabetic neuropathy in the streptozotocin-induced type 1 diabetic male rat? Can exercise and insulin-like growth factor 1 (IGF-I) treatment improve the diabetes-related decrease in angiogenesis in sciatic nerve in these animals? What is the main finding and its importance? Levels of VEGF-A, TSP-1 and NF-κB change in the sciatic nerve of diabetic rats and might mediate diabetic neuropathy. Treatment with IGF-I and exercise could increase angiogenesis in the diabetic rats by increasing VEGF-A and decreasing TSP-1 and NF-κB expression in the sciatic nerve. ABSTRACT Diabetic neuropathy is a severe complication of diabetes that affects 40-50% of diabetic people in the world. The aim of this study was to characterize alterations in angiogenesis and related molecular mediators in the sciatic nerve in diabetic conditions alone or in diabetes in combination with exercise and/or administration of insulin-like growth factor 1 (IGF-I). Forty male Wistar rats were assigned into one of five groups, namely control, diabetes, diabetes + exercise, diabetes + IGF-I and diabetes + exercise + IGF-I. Type 1 diabetes was induced by i.p. injection of streptozotocin (60 mg kg-1 ). After 30 days of treatment with exercise or IGF-I alone or in combination, diabetic neuropathy was evaluated with a hotplate, glycated haemoglobin was measured, angiogenesis was determined by immunostaining for PECAM-1/CD31, and expressions of vascular endothelial growth factor-A (VEGF-A), thrombospondin-1 (TSP-1) and nuclear factor-κB (NF-κB) were determined by enzyme-linked immunosorbent assay.After 4 weeks, the diabetes group showed a significant decrease in capillary density and VEGF-A levels, but a significant increase in glycated haemoglobin in blood, TSP-1 and NF-κB levels in the sciatic nerve compared with the control group, and these effects were ameliorated by exercise and IGF-I. However, simultaneous treatment of diabetic rats with IGF-I and exercise did not have any synergistic effects. These findings indicate that diabetes-induced neuropathy may be associated, in part, with decreased angiogenesis mediated by overproduction of TSP-1 and NF-κB, in addition to reduced production of VEGF-A. The findings also showed that exercise and IGF-I can reduce neuropathy, followed by increased angiogenesis, by changes in TSP-1, NF-κB and VEGF-A production levels.
Collapse
Affiliation(s)
- Ehsan Saboory
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Urmia, Zanjan, Iran
| | | | - Mahrokh Samadi
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Abdolrahman Biabanghard
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Chodari
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
43
|
The Role of NFκB in Healthy and Preeclamptic Placenta: Trophoblasts in the Spotlight. Int J Mol Sci 2020; 21:ijms21051775. [PMID: 32150832 PMCID: PMC7084575 DOI: 10.3390/ijms21051775] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/01/2023] Open
Abstract
The NFκB protein family regulates numerous pathways within the cell-including inflammation, hypoxia, angiogenesis and oxidative stress-all of which are implicated in placental development. The placenta is a critical organ that develops during pregnancy that primarily functions to supply and transport the nutrients required for fetal growth and development. Abnormal placental development can be observed in numerous disorders during pregnancy, including fetal growth restriction, miscarriage, and preeclampsia (PE). NFκB is highly expressed in the placentas of women with PE, however its contributions to the syndrome are not fully understood. In this review we discuss the molecular actions and related pathways of NFκB in the placenta and highlight areas of research that need attention.
Collapse
|
44
|
Masoumi-Dehghi S, Babashah S, Sadeghizadeh M. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways. J Cell Commun Signal 2020; 14:233-244. [PMID: 32034654 DOI: 10.1007/s12079-020-00548-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 01/20/2020] [Indexed: 12/21/2022] Open
Abstract
Paracrine signaling between tumor and surrounding stromal cells is critical for the maintenance of tumor microenvironment during ovarian cancer progression. Small extracellular vesicles (sEVs; exosomes in particular) are nano-sized vesicles secreted actively by many cells including tumor cells and are found to have fundamental roles in intercellular communication through shuttling functional RNAs. Although microRNAs (also called miRNAs or miRs), small non-coding RNAs regulating gene expression, are selectively accumulated in tumor sEVs and can mediate intercellular communication, the exact biological mechanisms underlying the functions of exosomal miRNAs in ovarian tumor angiogenesis remain unclear. In this study, sEVs were isolated from conditioned medium of the human ovarian carcinoma cell line SKOV-3 using ExoQuick Exosome Precipitation Solution, and characterized by scanning electron microscopy, dynamic light scattering, and immunoblotting. To elucidate the possible paracrine effects on ovarian tumor cell-derived sEVs (TD-sEVs), we investigated the angiogenesis-related signaling events triggered by TD-sEVs in endothelial cells. Due to the possible role in ovarian tumor pathogenesis, we focused on miR-141-3p which was detected to be enriched in TD-sEVs compared with their corresponding donor cells. We identified that sEV transfer of miR-141-3p considerably reduced the expression levels of cytokine-inducible suppressors of cytokine signaling (SOCS)-5 leading to up-regulated JAK-STAT3 pathway in endothelial cells. We also observed that sEV-shuttled miR-141-3p may up-regulate the expression of VEGFR-2 in endothelial cells which leads to promoting endothelial cell migration and angiogenesis. The putative role of miR-141-3p shuttled by TD-sEVs in regulating VEGFR-2 expression was demonstrated by the ability of anti-miR-141-3p to rescue the promoting effects of TD-sEVs on the expression of VEGFR-2 in endothelial cells. Our results also revealed that TD-sEVs trigger the intracellular reactive oxygen species (ROS)-dependent activation of NF-κB signaling in endothelial cells. Taken together, our findings propose a novel model in which sEV transfer of epithelial ovarian cancer-secreted miR-141-3p plays as a significant mediator of intercellular communication, promoting endothelial cell angiogenesis.
Collapse
Affiliation(s)
- Sajjad Masoumi-Dehghi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran.
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| |
Collapse
|
45
|
Wang R, Ma Y, Zhan S, Zhang G, Cao L, Zhang X, Shi T, Chen W. B7-H3 promotes colorectal cancer angiogenesis through activating the NF-κB pathway to induce VEGFA expression. Cell Death Dis 2020; 11:55. [PMID: 31974361 PMCID: PMC6978425 DOI: 10.1038/s41419-020-2252-3] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Tumor angiogenesis is a hallmark of cancer and is involved in the tumorigenesis of solid tumors. B7-H3, an immune checkpoint molecule, plays critical roles in proliferation, metastasis and tumorigenesis in diverse tumors; however, little is known about the biological functions and molecular mechanism underlying B7-H3 in regulating colorectal cancer (CRC) angiogenesis. In this study, we first demonstrated that the expression of B7-H3 was significantly upregulated and was positively associated with platelet endothelial cell adhesion molecule-1 (CD31) level in tissue samples from patients with CRC. In addition, a series of in vitro and in vivo experiments showed that conditioned medium from B7-H3 knockdown CRC cells significantly inhibited the migration, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs), whereas overexpression of B7-H3 had the opposite effect. Furthermore, B7-H3 promoted tumor angiogenesis by upregulating VEGFA expression. Recombinant VEGFA abolished the inhibitory effects of conditioned medium from shB7-H3 CRC cells on HUVEC angiogenesis, while VEGFA siRNA or a VEGFA-neutralizing antibody reversed the effects of conditioned medium from B7-H3-overexpressing CRC cells on HUVEC angiogenesis. Moreover, we verified that B7-H3 upregulated VEGFA expression and angiogenesis by activating the NF-κB pathway. Collectively, our findings identify the B7-H3/NF-κB/VEGFA axis in promoting CRC angiogenesis, which serves as a promising approach for CRC treatment.
Collapse
Affiliation(s)
- Ruoqin Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
| | - Yanchao Ma
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Shenghua Zhan
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Lei Cao
- Jiangsu Key Laboratory of Gastrointestinal tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Tongguo Shi
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China. .,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
| |
Collapse
|
46
|
Huang LY, Yen IC, Tsai WC, Lee SY. Rhodiola crenulata Suppresses High Glucose-Induced Matrix Metalloproteinase Expression and Inflammatory Responses by Inhibiting ROS-Related HMGB1-TLR4 Signaling in Endothelial Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:91-105. [DOI: 10.1142/s0192415x20500056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Rhodiola crenulata, a popular folk medicine for anti-altitude sickness in Tibet, has been shown to have protective effects against high glucose (HG)-induced endothelial cell dysfunction in human umbilical vein endothelial cells (HUVECs). However, its mechanisms of action are unclear. Here, we aimed to examine the effects and the mechanisms of action of Rhodiola crenulata extract (RCE) on matrix metalloproteinases (MMPs) and inflammatory responses under HG conditions. HUVECs were pretreated with RCE or untreated and then exposed to 33[Formula: see text]mM glucose medium for 24[Formula: see text]h. The levels of oxidative stress markers, MMPs, endogenous tissue inhibitors of MMPs (TIMPs), and adhesion molecules were determined. Zymography assays were also carried out. We found that RCE significantly decreased HG-induced increases in reactive oxygen species (ROS) and activation of MAPK and NF-[Formula: see text]B pathways. In addition, RCE not only significantly reduced the expression and activities of MMPs but also upregulated TIMP protein levels. Consistently, HG-induced activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response protein (MyD88) signaling pathway, intracellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), and high mobility group box 1 (HMGB1) as well as endothelial cell apoptosis was inhibited by RCE treatment. RCE exerts protective effects on endothelial cells against HG insult, partially by suppressing the HMGB1/TLR4 axis. These findings indicate that Rhodiola crenulata may be a potential therapeutic agent for diabetes-associated vascular diseases.
Collapse
Affiliation(s)
- Li-Yen Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Cheng Tsai
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
47
|
He Y, Wu Z, Qiu C, Wang X, Xiang Y, Lu T, He Y, Shang T, Zhu Q, Wang X, Zeng Q, Zhang H, Li D. Long non-coding RNA GAPLINC promotes angiogenesis by regulating miR-211 under hypoxia in human umbilical vein endothelial cells. J Cell Mol Med 2019; 23:8090-8100. [PMID: 31589383 PMCID: PMC6850972 DOI: 10.1111/jcmm.14678] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/19/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022] Open
Abstract
In this study, we investigated the role of a long non-coding RNA GAPLINC in angiogenesis using human umbilical vein endothelial cells (HUVEC). We found that hypoxia and hypoxia-inducible factor 1α (HIF-1α) increased the expression of GAPLINC in HUVEC cells. Moreover, GAPLINC overexpression down-regulated miR-211 and up-regulated Bcl2 protein expression. Further rescue experiments confirmed that hypoxia directly increased GAPLINC expression. GAPLINC overexpression also increased cell migration and vessel formation which promoted angiogenesis, and these changes were attributed to the increased expression of vascular endothelial growth factor receptors (VEGFR) and delta-like canonical notch ligand 4 (DLL4) receptors. Finally, we demonstrated that GAPLINC promotes vessel formation and migration by regulating MAPK and NF-kB signalling pathways. Taken together, these findings comprehensively demonstrate that overexpression of GAPLINC increases HUVEC cells angiogenesis under hypoxia condition suggesting that GAPLINC can be a potential target for critical limb ischaemia (CLI) treatment.
Collapse
Affiliation(s)
- Yangyan He
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Ziheng Wu
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Chenyang Qiu
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Xiaohui Wang
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Yilang Xiang
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Tian Lu
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Yunjun He
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Tao Shang
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Qianqian Zhu
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Xun Wang
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Qinglong Zeng
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Hongkun Zhang
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| | - Donglin Li
- Department of Vascular SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHang ZhouChina
| |
Collapse
|
48
|
Ye LJ, Zhou XC, Yin XJ, Shang Y, Xiao Y, Jiang YL, Wen XX. CARD9 downregulation suppresses the growth of oral squamous cell carcinoma by regulating NF-κB. Oral Dis 2019; 25:1886-1896. [PMID: 31306536 DOI: 10.1111/odi.13157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To discover the expression pattern and potential underlying mechanism of the caspase recruitment domain-containing protein 9 (CARD9) in oral squamous cell carcinoma (OSCC). METHODS Caspase recruitment domain-containing protein 9 expression was detected by qRT-PCR and Western blot in OSCC tissues and cells, and OSCC (CGHNC9 and OECM-1) cell lines were divided into control, NC siRNA, and CARD9 siRNA groups. Then, MTT, flow cytometry, wound-healing, and Transwell assays were carried out to determine the changes in cellular biological characteristics. Immunoblot assay was performed for the expressions of NF-κB pathway. Finally, we constructed the xenograft models in nude mice to validate the in vivo effect of CARD9 siRNA on OSCC cell growth. RESULTS Caspase recruitment domain-containing protein 9 was upregulated in both OSCC tissues and cells, exhibiting a close relation with major clinicopathological features of OSCC patients. Transfection of CARD9 siRNA inhibited the proliferation, migration, and invasion of OSCC cells with the enhanced cell apoptosis, and meanwhile, CARD9, p-p65/p65, p-IKKα/IKKα, and p-IkBα/IkBα were downregulated. The tumor formation assay on nude mice also suggested that CARD9 siRNA might block the in vivo growth of OSCC cells. CONCLUSION Caspase recruitment domain-containing protein 9 suppression results in the upregulation of NF-κB pathway with suppressed proliferation, migration, and invasion of OSCC cells and facilitates the apoptosis.
Collapse
Affiliation(s)
- Li-Jun Ye
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Xin-Chun Zhou
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Xiao-Jia Yin
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Yu Shang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Yue Xiao
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Yu-Ling Jiang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Xin-Xuan Wen
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| |
Collapse
|
49
|
Lodola F, Rosti V, Tullii G, Desii A, Tapella L, Catarsi P, Lim D, Moccia F, Antognazza MR. Conjugated polymers optically regulate the fate of endothelial colony-forming cells. SCIENCE ADVANCES 2019; 5:eaav4620. [PMID: 31598549 PMCID: PMC6764832 DOI: 10.1126/sciadv.aav4620] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/04/2019] [Indexed: 05/02/2023]
Abstract
The control of stem and progenitor cell fate is emerging as a compelling urgency for regenerative medicine. Here, we propose a innovative strategy to gain optical control of endothelial colony-forming cell fate, which represents the only known truly endothelial precursor showing robust in vitro proliferation and overwhelming vessel formation in vivo. We combine conjugated polymers, used as photo-actuators, with the advantages offered by optical stimulation over current electromechanical and chemical stimulation approaches. Light modulation provides unprecedented spatial and temporal resolution, permitting at the same time lower invasiveness and higher selectivity. We demonstrate that polymer-mediated optical excitation induces a robust enhancement of proliferation and lumen formation in vitro. We identify the underlying biophysical pathway as due to light-induced activation of TRPV1 channel. Altogether, our results represent an effective way to induce angiogenesis in vitro, which represents the proof of principle to improve the outcome of autologous cell-based therapy in vivo.
Collapse
Affiliation(s)
- F. Lodola
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy
- Corresponding author. (F.L.); (M.R.A.)
| | - V. Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - G. Tullii
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza L. Da Vinci 32, 20133 Milano, Italy
| | - A. Desii
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy
| | - L. Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro,” Novara, Italy
| | - P. Catarsi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - D. Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro,” Novara, Italy
| | - F. Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - M. R. Antognazza
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy
- Corresponding author. (F.L.); (M.R.A.)
| |
Collapse
|
50
|
Characterization of Novel Murine and Human PDAC Cell Models: Identifying the Role of Intestine Specific Homeobox Gene ISX in Hypoxia and Disease Progression. Transl Oncol 2019; 12:1056-1071. [PMID: 31174057 PMCID: PMC6556561 DOI: 10.1016/j.tranon.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 11/23/2022] Open
Abstract
Therapy failure and metastasis-associated mortality are stumbling blocks in the management of PDAC in patients. Failure of therapy is associated to intense hypoxic conditions of tumors. To develop effective therapies, a complete understanding of hypoxia-associated changes in genetic landscape of tumors during disease progression is needed. Because artificially immortalized cell lines do not rightly represent the disease progression, studying genetics of tumors in spontaneous models is warranted. In the current study, we generated a spectrum of spontaneous human (UM-PDC1; UM-PDC2) and murine (HI-PanL, HI-PancI, HI-PanM) models representing localized, invasive, and metastatic PDAC from a patient and transgenic mice (K-rasG12D/Pdxcre/Ink4a/p16-/). These spontaneous models grow vigorously under hypoxia and exhibit activated K-ras signaling, progressive loss of PTEN, and tumorigenicity in vivo. Whereas UM-PDC1 form localized tumors, the UM-PDC2 metastasize to lungs in mice. In an order of progression, these models exhibit genomic instability marked by gross chromosomal rearrangements, centrosome-number variations, Aurora-kinase/H2AX colocalization, loss of primary cilia, and α-tubulin acetylation. The RNA sequencing of hypoxic models followed by qRT-PCR validation and gene-set enrichment identified Intestine-Specific Homeobox factor (ISX)–driven molecular pathway as an indicator PDAC aggressivness. TCGA-PAAD clinical data analysis showed high ISX expression correlation to poor survival of PDAC patients, particularly women. The functional studies showed ISX as a regulator of i) invasiveness and migratory potential and ii) VEGF, MMP2, and NFκB activation in PDAC cells. We suggest that ISX is a potential druggable target and newly developed spontaneous cell models are valuable tools for studying mechanism and testing therapies for PDAC.
Collapse
|