1
|
da Rocha VP, Mansano BSDM, Dos Santos CFC, Teixeira ILA, de Oliveira HA, Vieira SS, Antonio EL, Izar MCDO, Fonseca FAH, Serra AJ. How long does the biological effect of a red light-emitting diode last on adipose-derived mesenchymal stem cells? Photochem Photobiol 2025; 101:206-214. [PMID: 38888236 DOI: 10.1111/php.13983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
This research investigated the duration of the influence of red light-emitting diodes (LED, 630 nm; output power: 2452.5 mW; laser beam: 163.5 cm2; irradiance: 15 mW/cm2; radiant exposure: 4 J/cm2) on different periods after irradiation (6, 12, 24, 48, and 72 h) on adipose-derived mesenchymal stem cells' (AdMSCs) metabolism and paracrine factors. AdMSCs were irradiated three times every 48 h. Twenty-four hours after the last irradiation, there was a higher MTT absorbance, followed by a decrease after 48 h. The cells' secretome showed increased levels of IL-6 and VEGF after 12 and 24 h, but this was reversed after 48 h. Additionally, LED irradiation resulted in higher levels of nitrite and did not affect oxidative stress markers. LED irradiation had significant effects on AdMSCs after 24 h compared to other groups and its control group.
Collapse
Affiliation(s)
- Vitor Pocani da Rocha
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | | | | | - Stella Sousa Vieira
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | - Andrey Jorge Serra
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| |
Collapse
|
2
|
Li S, Ye X, Wen X, Yang X, Wang L, Gao K, Xiao H, Jiang Z. Arginine and its metabolites stimulate proliferation, differentiation, and physiological function of porcine trophoblast cells through β-catenin and mTOR pathways. BMC Vet Res 2024; 20:167. [PMID: 38689278 PMCID: PMC11062007 DOI: 10.1186/s12917-024-04023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Arginine, which is metabolized into ornithine, proline, and nitric oxide, plays an important role in embryonic development. The present study was conducted to investigate the molecular mechanism of arginine in proliferation, differentiation, and physiological function of porcine trophoblast cells (pTr2) through metabolic pathways. The results showed that arginine significantly increased cell viability (P < 0.05). The addition of arginine had a quadratic tendency to increase the content of progesterone (P = 0.06) and protein synthesis rate (P = 0.03), in which the maximum protein synthesis rate was observed at 0.4 mM arginine. Arginine quadratically increased (P < 0.05) the intracellular contents of spermine, spermidine and putrescine, as well as linearly increased (P < 0.05) the intracellular content of NO in a dose-dependent manner. Arginine showed a quadratic tendency to increase the content of putrescine (P = 0.07) and a linear tendency to increase NO content (P = 0.09) in cell supernatant. Moreover, increasing arginine activated (P < 0.05) the mRNA expressions for ARG, ODC, iNOS and PCNA. Furthermore, inhibitors of arginine metabolism (L-NMMA and DFMO) both inhibited cell proliferation, while addition of its metabolites (NO and putrescine) promoted the cell proliferation and cell cycle, the mRNA expressions of PCNA, EGF and IGF-1, and increased (P < 0.05) cellular protein synthesis rate, as well as estradiol and hCG secretion (P < 0.05). In conclusion, our results suggested that arginine could promote cell proliferation and physiological function by regulating the metabolic pathway. Further studies showed that arginine and its metabolites modulate cell function mainly through β-catenin and mTOR pathways.
Collapse
Affiliation(s)
- Shuai Li
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Xiangyang Ye
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Xiaolu Wen
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Xuefen Yang
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Li Wang
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Kaiguo Gao
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China.
| | - Hao Xiao
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China.
| | - Zongyong Jiang
- Institute of Animal Science, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| |
Collapse
|
3
|
Kang SU, Kim HJ, Ma S, Oh DY, Jang JY, Seo C, Lee YS, Kim CH. Liquid plasma promotes angiogenesis through upregulation of endothelial nitric oxide synthase-induced extracellular matrix metabolism: potential applications of liquid plasma for vascular injuries. Cell Commun Signal 2024; 22:138. [PMID: 38374138 PMCID: PMC10875778 DOI: 10.1186/s12964-023-01412-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/25/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Applications of nonthermal plasma have expanded beyond the biomedical field to include antibacterial, anti-inflammatory, wound healing, and tissue regeneration. Plasma enhances epithelial cell repair; however, the potential damage to deep tissues and vascular structures remains under investigation. RESULT This study assessed whether liquid plasma (LP) increased nitric oxide (NO) production in human umbilical vein endothelial cells by modulating endothelial NO synthase (eNOS) phosphorylation and potential signaling pathways. First, we developed a liquid plasma product and confirmed the angiogenic effect of LP using the Matrigel plug assay. We found that the NO content increased in plasma-treated water. NO in plasma-treated water promoted cell migration and angiogenesis in scratch and tube formation assays via vascular endothelial growth factor mRNA expression. In addition to endothelial cell proliferation and migration, LP influenced extracellular matrix metabolism and matrix metalloproteinase activity. These effects were abolished by treatment with NG-L-monomethyl arginine, a specific inhibitor of NO synthase. Furthermore, we investigated the signaling pathways mediating the phosphorylation and activation of eNOS in LP-treated cells and the role of LKB1-adenosine monophosphate-activated protein kinase in signaling. Downregulation of adenosine monophosphate-activated protein kinase by siRNA partially inhibited LP-induced eNOS phosphorylation, angiogenesis, and migration. CONCLUSION The present study suggests that LP treatment may be a novel strategy for promoting angiogenesis in vascular damage. Video Abstract.
Collapse
Affiliation(s)
- Sung Un Kang
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Haeng Jun Kim
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Sukhwal Ma
- Medical Accelerator Research Team, Korea Institute of Radiological & Medical Sciences (KIRAMS), 75 Nowonro, Nowon-gu, Seoul, 01812, South Korea
| | - Doo-Yi Oh
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Chorong Seo
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Yun Sang Lee
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, Department of Molecular Science and Technology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-Gu, Suwon, 443-380, Republic of Korea.
| |
Collapse
|
4
|
Mansano BSDM, da Rocha VP, Teixeira ILA, de Oliveira HA, Vieira SS, Antonio EL, Tucci PJF, Serra AJ. Light-emitting Diode Can Enhance the Metabolism and Paracrine Action of Mesenchymal Stem Cells. Photochem Photobiol 2023; 99:1420-1428. [PMID: 36807286 DOI: 10.1111/php.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
This study investigated the influence of red light-emitting diodes (LED, 630 nm) on different irradiation parameters and the number of applications on mesenchymal stem cells derived from adipose tissue (AdMSCs) metabolism and paracrine factors. The AdMSCs were irradiated with a LEDbox device (output power: 2452.5 mW; laser beam: 163.5 cm2 ; irradiance: 15 mW cm-2 ) using radiant exposures of 0.5, 2, and 4 J cm-2 , respectively. AdMSCs were irradiated once or every 48 h up to three irradiations. All molecular analyses were performed 24 h after the last irradiation. LED did not induce changes in cell count, DNA damage, and oxidative stress. A significant repercussion of the LED has been noticed after three irradiations with 4 J cm-2 . AdMSCs had higher levels of IL-6, IGF-1, and NOx index. A higher ATP content and MMT/Resazurin assay were identified in AdMSCs irradiated three times with 4 J cm-2 . Mitochondrial basal respiration, maximal respiration and proton leak under metabolic stress were reduced by 0.5 and 2 J cm-2 irradiations. These data showed that three LED irradiations with 4 J cm-2 may be a suitable parameter for future AdMSCs therapy because of its improved metabolic activity, ATP content, and IL-6, IGF-1, and nitric oxide secretion.
Collapse
Affiliation(s)
| | - Vitor Pocani da Rocha
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | - Stella Souza Vieira
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
- Base Hospital Foundation, Medicine School of São José do Rio Preto, Sao Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | - Andrey Jorge Serra
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| |
Collapse
|
5
|
Stenhouse C, Bazer FW, Ashworth CJ. Sexual dimorphism in placental development and function: Comparative physiology with an emphasis on the pig. Mol Reprod Dev 2023; 90:684-696. [PMID: 35466463 DOI: 10.1002/mrd.23573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022]
Abstract
Across mammalian species, it has been demonstrated that sex influences birth weight, with males being heavier than females; a characteristic that can be observed from early gestation. Male piglets are more likely to be stillborn and have greater preweaning mortality than their female littermates, despite the additional maternal investment into male fetal growth. Given the conserved nature of the genome between the sexes, it is hypothesized that these developmental differences between males and females are most likely orchestrated by differential placental adaptation. This review summarizes the current understanding of fetal sex-specific differences in placental and endometrial structure and function, with an emphasis on pathways found to be differentially regulated in the pig including angiogenesis, apoptosis, and proliferation. Given the importance of piglet sex in agricultural enterprises, and the potential for skewed litter sex ratios, it is imperative to improve understanding of the relationship between fetal sex and molecular signaling in both the placenta and endometria across gestation.
Collapse
Affiliation(s)
- Claire Stenhouse
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Cheryl J Ashworth
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
6
|
Wei Y, Jiang H, Chai C, Liu P, Qian M, Sun N, Gao M, Zu H, Yu Y, Ji G, Zhang Y, Yang S, He J, Cheng J, Tian J, Zhao Q. Endothelium-Mimetic Surface Modification Improves Antithrombogenicity and Enhances Patency of Vascular Grafts in Rats and Pigs. JACC Basic Transl Sci 2023; 8:843-861. [PMID: 37547067 PMCID: PMC10401295 DOI: 10.1016/j.jacbts.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 08/08/2023]
Abstract
We first identified thrombomodulin (TM) and endothelial nitric oxide (NO) synthase as key factors for the antithrombogenic function of the endothelium in human atherosclerotic carotid arteries. Then, recombinant TM and an engineered galactosidase responsible for the conversion of an exogenous NO prodrug were immobilized on the surface of the vascular grafts. Surface modification by TM and NO cooperatively enhanced the antithrombogenicity and patency of vascular grafts. Importantly, we found that the combination of TM and NO also promoted endothelialization, whereas it reduced adverse intimal hyperplasia, which is critical for the maintenance of vascular homeostasis, as confirmed in rat and pig models.
Collapse
Affiliation(s)
- Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Huan Jiang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Chao Chai
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Pei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Na Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia (Ministry of Education), Harbin, China
| | - Man Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Honglin Zu
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yongquan Yu
- Department of Radiology, Weihai Central Hospital, Weihai, China
| | - Guangbo Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Yating Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Sen Yang
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ju He
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jiansong Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia (Ministry of Education), Harbin, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
7
|
Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022; 125:4-9. [PMID: 35414477 PMCID: PMC10947607 DOI: 10.1016/j.placenta.2022.03.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 01/16/2023]
Abstract
Pregnancy complications adversely impact both mother and/or fetus throughout the lifespan. Fetal growth restriction (FGR) occurs when a fetus fails to reach their intrauterine potential for growth, it is the second highest leading cause of infant mortality, and leads to increased risk of developing non-communicable diseases in later life due 'fetal programming'. Abnormal placental development, growth and/or function underlies approximately 75% of FGR cases and there is currently no treatment save delivery, often prematurely. We previously demonstrated in a murine model of FGR that nanoparticle mediated, intra-placental human IGF-1 gene therapy maintains normal fetal growth. Multiple models of FGR currently exist reflecting the etiologies of human FGR and have been used by us and others to investigate the development of in utero therapeutics as discussed here. In addition to the in vivo models discussed herein, utilizing human models including in vitro (Choriocarcinoma cell lines and primary trophoblasts) and ex vivo (term villous fragments and placenta cotyledon perfusion) we have demonstrated robust nanoparticle uptake, transgene expression, nutrient transporter regulation without transfer to the fetus. For translational gene therapy application in the human placenta, there are multiple avenues that require investigation including syncytial uptake from the maternal circulation, transgene expression, functionality and longevity of treatment, impact of treatment on the mother and developing fetus. The potential impact of treating the placenta during gestation is high, wide-ranging across pregnancy complications, and may offer reduced risk of developing associated cardio-metabolic diseases in later life impacting at both an individual and societal level.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States.
| |
Collapse
|
8
|
Wang F, Qin K, Wang K, Wang H, Liu Q, Qian M, Chen S, Sun Y, Hou J, Wei Y, Hu Y, Li Z, Xu Q, Zhao Q. Nitric oxide improves regeneration and prevents calcification in bio-hybrid vascular grafts via regulation of vascular stem/progenitor cells. Cell Rep 2022; 39:110981. [PMID: 35732119 DOI: 10.1016/j.celrep.2022.110981] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 04/29/2022] [Accepted: 05/28/2022] [Indexed: 11/18/2022] Open
Abstract
Vascular bypass surgery continues to use autologous grafts and often suffers from a shortage of donor grafts. Decellularized xenografts derived from porcine veins provide a promising candidate because of their abundant availability and low immunogenicity. Unfortunately, transplantation outcomes are far from satisfactory because of insufficient regeneration and adverse pathologic remodeling. Herein, a nitrate-functionalized prosthesis has been incorporated into a decellularized porcine vein graft to fabricate a bio-hybrid vascular graft with local delivery of nitric oxide (NO). Exogenous NO efficiently promotes vascular regeneration and attenuates intimal hyperplasia and vascular calcification in both rabbit and mouse models. The underlying mechanism was investigated using a Sca1 2A-CreER; Rosa-RFP genetic-lineage-tracing mouse model that reveals that Sca1+ stem/progenitor cells (SPCs) are major contributors to vascular regeneration and remodeling, and NO plays a critical role in regulating SPC fate. These results support the translational potential of this off-the-shelf vascular graft.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou 256600, China
| | - Kang Qin
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - He Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qi Liu
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shang Chen
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yijin Sun
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jingli Hou
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanhua Hu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
9
|
Xu Y, Jiang X, Niu C, Yang S, Xiao X, Huang Z, Feng L. Preparation and Assessment of Nitric Oxide‐releasing Small‐diameter Collagen‐based Vascular Graft for Vascular Regeneration Application. MACROMOLECULAR MATERIALS AND ENGINEERING 2022. [DOI: 10.1002/mame.202100862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Yue Xu
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Xia Jiang
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Chuan Niu
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Shaojie Yang
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Xiong Xiao
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Ziwei Huang
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| | - Li Feng
- Regenerative Medicine Research Center West China Hospital Sichuan University Chengdu People's Republic of China
| |
Collapse
|
10
|
Krause BJ. Novel insights for the role of nitric oxide in placental vascular function during and beyond pregnancy. J Cell Physiol 2021; 236:7984-7999. [PMID: 34121195 DOI: 10.1002/jcp.30470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023]
Abstract
More than 30 years have passed since endothelial nitric oxide synthesis was described using the umbilical artery and vein endothelium. That seminal report set the cornerstone for unveiling the molecular aspects of endothelial function. In parallel, the understanding of placental physiology has gained growing interest, due to its crucial role in intrauterine development, with considerable long-term health consequences. This review discusses the evidence for nitric oxide (NO) as a critical player of placental development and function, with a special focus on endothelial nitric oxide synthase (eNOS) vascular effects. Also, the regulation of eNOS-dependent vascular responses in normal pregnancy and pregnancy-related diseases and their impact on prenatal and postnatal vascular health are discussed. Recent and compelling evidence has reinforced that eNOS regulation results from a complex network of processes, with novel data concerning mechanisms such as mechano-sensing, epigenetic, posttranslational modifications, and the expression of NO- and l-arginine-related pathways. In this regard, most of these mechanisms are expressed in an arterial-venous-specific manner and reflect traits of the fetal systemic circulation. Several studies using umbilical endothelial cells are not aimed to understand placental function but general endothelial function, reinforcing the influence of the placenta on general knowledge in physiology.
Collapse
Affiliation(s)
- Bernardo J Krause
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| |
Collapse
|
11
|
Massimiani M, Tiralongo GM, Salvi S, Fruci S, Lacconi V, La Civita F, Mancini M, Stuhlmann H, Valensise H, Campagnolo L. Treatment of pregnancies complicated by intrauterine growth restriction with nitric oxide donors increases placental expression of Epidermal Growth Factor-Like Domain 7 and improves fetal growth: A pilot study. Transl Res 2021; 228:28-41. [PMID: 32784003 DOI: 10.1016/j.trsl.2020.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 11/25/2022]
Abstract
Intrauterine growth restriction (IUGR) is a pathological condition of pregnancy with high perinatal mortality and morbidity, characterized by inadequate fetal growth associated to altered maternal hemodynamics with impaired uteroplacental blood flow and placental insufficiency. To date, iatrogenic premature delivery remains the elective therapeutic strategy. However, in recent years the possibility of a therapeutic approach with vasodilators and myorelaxants, such as nitric oxide (NO) donors, has gained interest. NO controls many endothelial cell functions, including angiogenesis and vascular permeability, by regulating the expression of angiogenic factors, such as Vascular Endothelial Growth Factor. In the present study, we investigated if treatment of pregnancies complicated by IUGR with NO donors affects the expression of Epidermal Growth Factor-Like Domain 7 (EGFL7), a secreted endothelial factor, previously demonstrated to be expressed by both endothelial and trophoblast cells and involved in proper placental development. NO donor treatment induced placental levels of EGFL7 and, in association with oral fluids, significantly improved fetal growth. Ex vivo experiments confirmed that NO donors increased expression and secretion of EGFL7 by villous explants. To specifically investigate the potential response of trophoblast cells to NO, we treated HTR8-sVneo cells with NO donors and observed induction of EGFL7 expression. Altogether, our findings indicate that NO induces endothelial and trophoblast expression of EGFL7 in the placenta and improves fetal growth, suggesting a correlation between placental levels of EGFL7 and pregnancy outcome.
Collapse
Affiliation(s)
- Micol Massimiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Saint Camillus International University of Health Sciences, Rome, Italy
| | - Grazia M Tiralongo
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy
| | - Silvia Salvi
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, UOC di Patologia Ostetrica, Fondazione Policlinico Universitario "A Gemelli" IRCCS, Rome, Italy
| | | | - Valentina Lacconi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Fabio La Civita
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Marta Mancini
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York
| | - Herbert Valensise
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy; Obstetrics and Gynecology Unit, Department of Surgical Sciences, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
12
|
Nitric oxide: Is it the culprit for the continued expansion of keloids? Eur J Pharmacol 2019; 854:282-288. [PMID: 31034822 DOI: 10.1016/j.ejphar.2019.04.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/19/2019] [Accepted: 04/25/2019] [Indexed: 11/22/2022]
Abstract
Keloids are characterized by excessive proliferation of fibroblasts and invasion of surrounding healthy skin. High levels of Nitric Oxide (NO) are thought to be the crucial factor within the micro-environment in promoting keloid formation. However, the effects and mechanisms of NO on the proliferation of Keloid Fibroblasts (KDFs) remain unclear. In this study, we investigated the effect of NO on KDFs proliferation by Sodium Nitroprusside (SNP), an NO donor. Our results show that SNP significantly enhanced KDFs proliferation. Moreover, with prolonged treatment with SNP after cell confluence, the growth of KDFs escape contact inhibition and experience significant pile up growth. Furthermore, PTIO, an NO scavenger, attenuated SNP-enhanced cell proliferation effectively. The mechanism involved in SNP-induced KDFs proliferation was soluble Guanylyl Cyclase (sGC) and cGMP independent. ODQ, a specific sGC inhibitor, failed to suppress SNP-enhanced KDFs proliferation. 8-Bromo-c GMP, a cell-permeable cGMP analogue, could not stimulate KDFs proliferation. Erk and Akt provide important signaling for cell growth. U0126 and LY294002, inhibitors of Erk and Akt respectively, block SNP-enhanced KDFs proliferation effectively. As expected, a Western blot showed that SNP up-regulated the phosphorylation levels of Erk and Akt. Moreover, it decreased the expression of p27, a cell cycle inhibitor. Our results reveal that SNP induced KDFs proliferation and loss contact inhibition led to pile up growth via activation of the Erk and Akt pathways, as well as a decreased expression of p27. Thus, we speculate that the pathological feature of continuous expansion in keloids is caused by NO-induced KDFs sustained growth.
Collapse
|
13
|
Stenhouse C, Hogg CO, Ashworth CJ. Association of foetal size and sex with porcine foeto-maternal interface integrin expression. Reproduction 2019; 157:317-328. [PMID: 30650060 PMCID: PMC6391912 DOI: 10.1530/rep-18-0520] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/16/2019] [Indexed: 12/31/2022]
Abstract
Integrins regulate adhesion at the foeto-maternal interface by interacting with secreted phosphoprotein 1 (SPP1) and fibronectin (FN). It is hypothesised that impaired foetal growth of ‘runt’ piglets is linked to altered integrin signalling at the foeto-maternal interface. Placental and endometrial samples associated with the lightest and closest to mean litter weight (CTMLW) (gestational day (GD18, 30, 45, 60 and 90), of both sex (GD30, 45, 60 and 90) (n = 5–8 litters/GD), Large White × Landrace conceptuses or foetuses were obtained. The mRNA expression of the integrin subunits (ITG) ITGA2, ITGAV, ITGB1, ITGB3, ITGB5, ITGB6, ITGB8, SPP1 and FN was quantified by qPCR. Temporal changes in mRNA expression were observed, with different profiles in the two tissues. Endometrial ITGB1 (P ≤ 0.05, GD45) and SPP1 (P ≤ 0.05, all GD combined and GD60) expression was decreased in samples supplying the lightest compared to the CTMLW foetuses. Placentas supplying female foetuses had decreased expression of ITGB6 (GD45, P ≤ 0.05) and FN (GD90, P ≤ 0.05) compared to those supplying male foetuses. Endometrial samples supplying females had increased ITGB3 (P ≤ 0.05, GD60) and FN (P ≤ 0.05, GD30) expression and decreased SPP1 (P ≤ 0.05, GD60) expression compared to male foetuses. Correlations between mean within-gilt mRNA expression and percentage prenatal survival, number of live foetuses or conceptuses and percentage male foetuses were observed. This study has highlighted novel and dynamic associations between foetal size, sex and integrin subunit mRNA expression at the porcine foeto-maternal interface. Further studies should be performed to improve the understanding of the mechanisms behind these novel findings.
Collapse
Affiliation(s)
- Claire Stenhouse
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Charis O Hogg
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Cheryl J Ashworth
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
14
|
Zhang HH, Lechuga TJ, Chen Y, Yang Y, Huang L, Chen DB. Quantitative Proteomics Analysis of VEGF-Responsive Endothelial Protein S-Nitrosylation Using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and LC-MS/MS. Biol Reprod 2016; 94:114. [PMID: 27075618 PMCID: PMC4939742 DOI: 10.1095/biolreprod.116.139337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/08/2016] [Accepted: 04/06/2016] [Indexed: 12/20/2022] Open
Abstract
Adduction of a nitric oxide moiety (NO•) to cysteine(s), termed S-nitrosylation (SNO), is a novel mechanism for NO to regulate protein function directly. However, the endothelial SNO-protein network that is affected by endogenous and exogenous NO is obscure. This study was designed to develop a quantitative proteomics approach using stable isotope labeling by amino acids in cell culture for comparing vascular endothelial growth factor (VEGFA)- and NO donor-responsive endothelial nitroso-proteomes. Primary placental endothelial cells were labeled with "light" (L-(12)C6 (14)N4-Arg and L-(12)C6 (14)N2-Lys) or "heavy" (L-(13)C6 (15)N4-Arg and L-(13)C6 (15)N2-Lys) amino acids. The light cells were treated with an NO donor nitrosoglutathione (GSNO, 1 mM) or VEGFA (10 ng/ml) for 30 min, while the heavy cells received vehicle as control. Equal amounts of cellular proteins from the light (GSNO or VEGFA treated) and heavy cells were mixed for labeling SNO-proteins by the biotin switch technique and then trypsin digested. Biotinylated SNO-peptides were purified for identifying SNO-proteins by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Ratios of light to heavy SNO-peptides were calculated for determining the changes of the VEGFA- and GSNO-responsive endothelial nitroso-proteomes. A total of 387 light/heavy pairs of SNO-peptides were identified, corresponding to 213 SNO-proteins that include 125 common and 27 VEGFA- and 61 GSNO-responsive SNO-proteins. The specific SNO-cysteine(s) in each SNO-protein were simultaneously identified. Pathway analysis revealed that SNO-proteins are involved in various endothelial functions, including proliferation, motility, metabolism, and protein synthesis. We collectively conclude that endogenous NO on VEGFA stimulation and exogenous NO from GSNO affect common and different SNO-protein networks, implicating SNO as a critical mechanism for VEGFA stimulation of angiogenesis.
Collapse
Affiliation(s)
- Hong-Hai Zhang
- Department of Obstetrics and Gynecology, University of California, Irvine, California
| | - Thomas J Lechuga
- Department of Obstetrics and Gynecology, University of California, Irvine, California
| | - Yuezhou Chen
- Department of Obstetrics and Gynecology, University of California, Irvine, California
| | - Yingying Yang
- Department of Biophysics and Physiology, University of California, Irvine, California
| | - Lan Huang
- Department of Biophysics and Physiology, University of California, Irvine, California
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California, Irvine, California
| |
Collapse
|
15
|
Chen DB, Zheng J. Regulation of placental angiogenesis. Microcirculation 2014; 21:15-25. [PMID: 23981199 DOI: 10.1111/micc.12093] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/21/2013] [Indexed: 12/13/2022]
Abstract
Ample interest has been evoked in using placental angiogenesis as a target for the development of diagnosis tools and potential therapeutics for pregnancy complications based on the knowledge of placental angiogenesis in normal and aberrant pregnancies. Although these goals are still far from reach, one would expect that two complementary processes should be balanced for therapeutic angiogenesis to be successful in restoring a mature and functional vascular network in the placenta in any pregnancy complication: (i) pro-angiogenic stimulation of new vessel growth and (ii) anti-angiogenic inhibition of vessel overgrowth. As the best model of physiological angiogenesis, investigations of placental angiogenesis provide critical insights not only for better understanding of normal placental endothelial biology but also for the development of diagnosis tools for pregnancy complications. Such investigations will potentially identify novel pro-angiogenic factors for therapeutic intervention for tissue damage in various obstetric complications or heart failure or anti-angiogenic factors to target on cancer or vision loss in which circulation needs to be constrained. This review summarizes the genetic and molecular aspects of normal placental angiogenesis as well as the signaling mechanisms by which the dominant angiogenic factor vascular endothelial growth factor regulates placental angiogenesis with a focus on placental endothelial cells.
Collapse
Affiliation(s)
- Dong-Bao Chen
- Department of Obstetrics & Gynecology, University of California, Irvine, California, USA
| | | |
Collapse
|
16
|
Zhang QY, Wang ZY, Wen F, Ren L, Li J, Teoh SH, Thian ES. Gelatin-siloxane nanoparticles to deliver nitric oxide for vascular cell regulation: synthesis, cytocompatibility, and cellular responses. J Biomed Mater Res A 2014; 103:929-38. [PMID: 24853642 DOI: 10.1002/jbm.a.35239] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/10/2014] [Accepted: 05/19/2014] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is an important mediator in cardiovascular system to regulate vascular tone and maintain tissue homeostasis. Its role in vascular cell regulation makes it promising to address the post-surgery restenosis problem. However, the application of NO is constrained by its high reactivity. Here, we developed a novel NO-releasing gelatin-siloxane nanoparticle (GS-NO NP) to deliver NO effectively for vascular cell regulation. Results showed that gelatin-siloxane nanoparticles (GS NPs) could be synthesized via sol-gel chemistry with a diameter of ∼200 nm. It could be modified into GS-NO NPs via S-nitrosothiol (RSNO) modification. The synthesized GS-NO NPs could release a total of ∼0.12 µmol/mg NO sustainably for 7 days following a first-order exponential profile. They showed not only excellent cytocompatibility, but also rapid intracellularization within 2 h. GS-NO NPs showed inhibition of human aortic smooth muscle cell (AoSMC) proliferation and promotion of human umbilical vein endothelial cell (HUVEC) proliferation in a dose-dependent manner, which is an important approach to prevent restenosis. With GS-NO NP dose at 100 µg/mL, the proliferation of AoSMCs could be slowed down whereas the growth of HUVECs was significantly promoted. We concluded that GS-NO NPs could have potential to be used as a promising nano-system to deliver NO for vascular cell regulation.
Collapse
Affiliation(s)
- Qin-Yuan Zhang
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
17
|
Fujiwara T, Kanazawa S, Ichibori R, Tanigawa T, Magome T, Shingaki K, Miyata S, Tohyama M, Hosokawa K. L-arginine stimulates fibroblast proliferation through the GPRC6A-ERK1/2 and PI3K/Akt pathway. PLoS One 2014; 9:e92168. [PMID: 24651445 PMCID: PMC3961283 DOI: 10.1371/journal.pone.0092168] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 02/19/2014] [Indexed: 12/12/2022] Open
Abstract
l-Arginine is considered a conditionally essential amino acid and has been shown to enhance wound healing. However, the molecular mechanisms through which arginine stimulates cutaneous wound repair remain unknown. Here, we evaluated the effects of arginine supplementation on fibroblast proliferation, which is a key process required for new tissue formation. We also sought to elucidate the signaling pathways involved in mediating the effects of arginine on fibroblasts by evaluation of extracellular signal-related kinase (ERK) 1/2 activation, which is important for cell growth, survival, and differentiation. Our data demonstrated that addition of 6 mM arginine significantly enhanced fibroblast proliferation, while arginine deprivation increased apoptosis, as observed by enhanced DNA fragmentation. In vitro kinase assays demonstrated that arginine supplementation activated ERK1/2, Akt, PKA and its downstream target, cAMP response element binding protein (CREB). Moreover, knockdown of GPRC6A using siRNA blocked fibroblast proliferation and decreased phosphorylation of ERK1/2, Akt and CREB. The present experiments demonstrated a critical role for the GPRC6A-ERK1/2 and PI3K/Akt signaling pathway in arginine-mediated fibroblast survival. Our findings provide novel mechanistic insights into the positive effects of arginine on wound healing.
Collapse
Affiliation(s)
- Takashi Fujiwara
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita-shi, Osaka, Japan
| | - Shigeyuki Kanazawa
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita-shi, Osaka, Japan
- * E-mail:
| | - Ryoko Ichibori
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita-shi, Osaka, Japan
| | - Tomoko Tanigawa
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita-shi, Osaka, Japan
| | - Takuya Magome
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita-shi, Osaka, Japan
| | - Kenta Shingaki
- Department of Research & Development Noevir Co., Ltd. Higashiomi, Shiga, Japan
| | - Shingo Miyata
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osakasayama, Osaka, Japan
| | - Masaya Tohyama
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osakasayama, Osaka, Japan
| | - Ko Hosokawa
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita-shi, Osaka, Japan
| |
Collapse
|
18
|
Belgorosky D, Langle Y, Cormick BPM, Colombo L, Sandes E, Eiján AM. Inhibition of nitric oxide is a good therapeutic target for bladder tumors that express iNOS. Nitric Oxide 2014; 36:11-8. [DOI: 10.1016/j.niox.2013.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/23/2013] [Accepted: 10/28/2013] [Indexed: 01/22/2023]
|
19
|
Zhao Q, Zhang J, Song L, Ji Q, Yao Y, Cui Y, Shen J, Wang PG, Kong D. Polysaccharide-based biomaterials with on-demand nitric oxide releasing property regulated by enzyme catalysis. Biomaterials 2013; 34:8450-8. [DOI: 10.1016/j.biomaterials.2013.07.045] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/12/2013] [Indexed: 12/21/2022]
|
20
|
Burke AJ, Sullivan FJ, Giles FJ, Glynn SA. The yin and yang of nitric oxide in cancer progression. Carcinogenesis 2013; 34:503-12. [PMID: 23354310 DOI: 10.1093/carcin/bgt034] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Nitric oxide (NO) is a short-lived, pleiotropic molecule that affects numerous critical functions in the body. Presently, there are markedly conflicting findings in the literature regarding NO and its role in carcinogenesis and tumor progression. NO has been shown to have dichotomous effects on cellular proliferation, apoptosis, migration, invasion, angiogenesis and many other important processes in cancer biology. It has been shown to be both pro- and antitumorigenic, depending on the concentration and the tumor microenvironment in question. NO is generated by three isoforms of NO synthase (NOS) that are widely expressed and sometimes upregulated in human tumors. Due to its vast array of physiological functions, it presents a huge challenge to researchers to discover its true potential in cancer biology and consequently, its use in anticancer therapies. In this study, we review the current knowledge in this area, with an emphasis placed on NO modulation as an anticancer therapy, focusing on NO-donating drugs and NOS inhibitors.
Collapse
Affiliation(s)
- Amy J Burke
- Prostate Cancer Institute, National University of Ireland Galway, Dublin, Ireland.
| | | | | | | |
Collapse
|
21
|
Holder MJ, Milward MR, Palin WM, Hadis MA, Cooper PR. Effects of red light-emitting diode irradiation on dental pulp cells. J Dent Res 2012; 91:961-6. [PMID: 22879579 DOI: 10.1177/0022034512456040] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Light irradiation activates a range of cellular processes in a variety of cell types, including stem cells, and can promote tissue repair. This study investigated the effects of light-emitting diode (LED) exposure on dental pulp cells (DPCs). Dose response analysis at 20-second intervals up to 120 seconds demonstrated that a LED array emitting 653-nm red light stimulated significantly increased cell growth at 3 and 7 days post-irradiation with 40 (149 mJ/cm(2)) and 60 (224 mJ/cm(2)) seconds of radiant exposure. Double-dosing cells at days 1 and 4 of a 7-day culture period with 60-second (224 mJ/cm(2)) LED exposure significantly increased cell growth compared with a single dosing regime. BrdU analysis demonstrated significantly increased proliferation rates associated with significantly increased ATP, nitric oxide (NO), and mitochondrial metabolic activity. LED-stimulated NO levels were not reduced by inhibition of NO-synthase activity. Light exposure also rescued the inhibition of mitochondrial dysfunction and increased levels of in vitro mineralization compared with control. Media exchange experiments indicated that autocrine signaling was not likely responsible for red-light-induced DPC activity. In conclusion, data analysis indicated that 653-nm LED irradiation promoted DPC responses relevant to tissue repair, and this is likely mediated by increased mitochondrial activity.
Collapse
Affiliation(s)
- M J Holder
- Oral Biology, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | | | | | | |
Collapse
|
22
|
Feng L, Liao WX, Luo Q, Zhang HH, Wang W, Zheng J, Chen DB. Caveolin-1 orchestrates fibroblast growth factor 2 signaling control of angiogenesis in placental artery endothelial cell caveolae. J Cell Physiol 2012; 227:2480-91. [PMID: 21830216 DOI: 10.1002/jcp.22984] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fibroblast growth factor (FGF) receptor 1 (FGFR1) protein was expressed as the long and short as well as some truncated forms in ovine fetoplacental artery ex vivo and in vitro. Upon FGF2 stimulation, both the long and short FGFR1s were tyrosine phosphorylated and the PI3K/AKT1 and ERK1/2 pathways were activated in a concentration- and time- dependent manner in ovine fetoplacental artery endothelial (oFPAE) cells. Blockade of the PI3K/AKT1 pathway attenuated FGF2-stimulated cell proliferation and migration as well as tube formation; blockade of the ERK1/2 pathway abolished FGF2-stimulated tube formation and partially inhibited cell proliferation and did not alter cell migration. Both AKT1 and ERK1/2 were co-fractionated with caveolin-1 and activated by FGF2 in the caveolae. Disruption of caveolae by methyl-β-cyclodextrin inhibited FGF2 activation of AKT1 and ERK1/2. FGFR1 was found in the caveolae where it physically binds to caveolin-1. FGF2 stimulated dissociation of FGFR1 from caveolin-1. Downregulation of caveolin-1 significantly attenuated the FGF2-induced activation of AKT1 and ERK1/2 and inhibited FGF2-induced cell proliferation, migration and tube formation in oFPAE cells. Pretreatment with a caveolin-1 scaffolding domain peptide to mimic caveolin-1 overexpression also inhibited these FGF2-induced angiogenic responses. These data demonstrate that caveolae function as a platform for regulating FGF2-induced angiogenesis through spatiotemporally compartmentalizing FGFR1 and the AKT1 and ERK1/2 signaling modules; the major caveolar structural protein caveolin-1 interacts with FGFR1 and paradoxically regulates FGF2-induced activation of PI3K/AKT1 and ERK1/2 pathways that coordinately regulate placental angiogenesis.
Collapse
Affiliation(s)
- Lin Feng
- Department of Obstetrics & Gynecology, University of California Irvine, Irvine, California 92697, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
During normal pregnancy, dramatically increased placental blood flow is critical for fetal growth and survival as well as neonatal birth weights and survivability. This increased blood flow results from angiogenesis, vasodilatation, and vascular remodeling. Locally produced growth factors including fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA) are key regulators of placental endothelial functions including cell proliferation, migration, and vasodilatation. However, the precise signaling mechanisms underlying such regulation in fetoplacental endothelium are less well defined, specifically with regard to the interactions amongst protein kinases (PKs), protein phosphatase, and nitric oxide (NO). Recently, we and other researchers have obtained solid evidence showing that different signaling mechanisms participate in FGF2- and VEGFA-regulated fetoplacental endothelial cell proliferation and migration as well as NO production. This review will briefly summarize currently available data on signaling mediating fetoplacental angiogenesis with a specific emphasis on PKs, ERK1/2, AKT1, and p38 MAPK and protein phosphatases, PPP2 and PPP3.
Collapse
Affiliation(s)
- Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715
- Address correspondence and reprint requests to: Jing Zheng, Ph.D., Departments of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin, PAB1 Meriter Hospital, 202 S Park St., Madison, WI 53715. Phone: (608) 417-6314 Fax: (608) 257-1304.
| |
Collapse
|
24
|
Kong X, Tan B, Yin Y, Gao H, Li X, Jaeger LA, Bazer FW, Wu G. L-Arginine stimulates the mTOR signaling pathway and protein synthesis in porcine trophectoderm cells. J Nutr Biochem 2011; 23:1178-83. [PMID: 22137265 DOI: 10.1016/j.jnutbio.2011.06.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/01/2011] [Accepted: 06/29/2011] [Indexed: 11/24/2022]
Abstract
Impairment of placental growth is a major factor contributing to intrauterine growth retardation (IUGR) in both human pregnancy and animal production. Results of recent studies indicate that administration of L-arginine (Arg) to gestating pigs or sheep with IUGR fetuses can enhance fetal growth. However, the underlying mechanisms are largely unknown. The present study tested the hypothesis that Arg stimulates the mammalian target of rapamycin (mTOR) signaling pathway and protein synthesis in porcine conceptus trophectoderm (pTr2) cells. The cells were cultured for 4 days in Arg-free Dulbecco's modified Eagle's Ham medium containing 10, 50, 100, 200, 350 or 500 μM Arg. Cell numbers, protein synthesis and degradation, as well as total and phosphorylated levels of mTOR, ribosomal protein S6 kinase 1 (p70S6K) and eukaryotic initiation factor 4E-binding protein-1 (4EBP1), were determined. The pTr2 cells exhibited time (0-6 days)- and Arg concentration (10-350 μM)-dependent increases in proliferation. Addition of 100 and 350 μM Arg to culture medium dose-dependently increased (a) protein synthesis and decreased protein degradation and (b) the abundance of total and phosphorylated mTOR, p70S6K and 4EBP1 proteins. Effects of 350 μM Arg on intracellular protein turnover were only modestly affected when nitric oxide synthesis was inhibited. Collectively, these results indicate a novel and important role for Arg in promoting growth of porcine placental cells largely via a nitric-oxide-independent pathway. Additionally, these findings help to explain beneficial effects of Arg supplementation on improving survival and growth of embryos/fetuses in mammals.
Collapse
Affiliation(s)
- Xiangfeng Kong
- Research Center for Healthy Breeding of Livestock and Poultry and Key Laboratory for Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, 410125 Hunan, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Krause B, Hanson M, Casanello P. Role of nitric oxide in placental vascular development and function. Placenta 2011; 32:797-805. [PMID: 21798594 PMCID: PMC3218217 DOI: 10.1016/j.placenta.2011.06.025] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 11/27/2022]
Abstract
Nitric oxide (NO) is one of the most pleiotropic signaling molecules at systemic and cellular levels, participating in vascular tone regulation, cellular respiration, proliferation, apoptosis and gene expression. Indeed NO actively participates in trophoblast invasion, placental development and represents the main vasodilator in this tissue. Despite the large number of studies addressing the role of NO in the placenta, its participation in placental vascular development and the effect of altered levels of NO on placental function remains to be clarified. This review draws a time-line of the participation of NO throughout placental vascular development, from the differentiation of vascular precursors to the consolidation of vascular function are considered. The influence of NO on cell types involved in the origin of the placental vasculature and the expression and function of the nitric oxide synthases (NOS) throughout pregnancy are described. The developmental processes involved in the placental vascular bed are considered, such as the participation of NO in placental vasculogenesis and angiogenesis through VEGF and Angiopoietin signaling molecules. The role of NO in vascular function once the placental vascular tree has developed, in normal pregnancy as well as in pregnancy-related diseases, is then discussed.
Collapse
Affiliation(s)
- B.J. Krause
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - M.A. Hanson
- Institute of Developmental Sciences, Academic Unit of Human Development & Health, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - P. Casanello
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
26
|
Necroptosis: an emerging form of programmed cell death. Crit Rev Oncol Hematol 2011; 82:249-58. [PMID: 21962882 DOI: 10.1016/j.critrevonc.2011.08.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 07/17/2011] [Accepted: 08/10/2011] [Indexed: 02/08/2023] Open
Abstract
Necrosis plays an important role in multiple physiological and pathological processes. Recently, a relatively new form of necrosis has been characterized as "necroptosis". Morphologically, necroptosis exhibits the features of necrosis; however, necroptosis exhibits a unique signaling pathway that requires the involvement of receptor interaction protein kinase 1 and 3 (RIP1 and RIP3) and can be specifically inhibited by necrostatins. Necroptosis has been found to contribute to the regulation of immune system, cancer development as well as cellular responses to multiple stresses. In this review, we will summarize the signaling pathway, biological effects and pathological significance of this specific form of programmed cell death.
Collapse
|
27
|
Kim J, Burghardt RC, Wu G, Johnson GA, Spencer TE, Bazer FW. Select Nutrients in the Ovine Uterine Lumen. IX. Differential Effects of Arginine, Leucine, Glutamine, and Glucose on Interferon Tau, Ornithine Decarboxylase, and Nitric Oxide Synthase in the Ovine Conceptus1. Biol Reprod 2011; 84:1139-47. [DOI: 10.1095/biolreprod.110.088153] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
28
|
Li SM, Zeng LW, Feng L, Chen DB. Rac1-dependent intracellular superoxide formation mediates vascular endothelial growth factor-induced placental angiogenesis in vitro. Endocrinology 2010; 151:5315-25. [PMID: 20844008 PMCID: PMC2954717 DOI: 10.1210/en.2010-0178] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vascular endothelial growth factor (VEGF) is one of the best characterized angiogenic factors controlling placental angiogenesis; however, how VEGF regulates placental angiogenesis has not yet completely understood. In this study, we found that all the components of assembling a functional NADPH oxidase (NOX2, p22(phox), p47(phox), p67(phox), and Rac1) are expressed in ovine fetoplacental artery endothelial cells (oFPAECs) in vitro and ex vivo. Treatment with VEGF (10 ng/ml) rapidly and transiently activated Rac1 in oFPAECs in vitro and increased Rac1 association with p67(phox) in 5 min. Intracellular superoxide formation began to significantly increase after 25-30 min of VEGF stimulation, which was mediated by both VEGFR1 and VEGFR2. VEGF also stimulated oFPAE cell proliferation and migration and enhanced the formation of tube-like structures on Matrigel matrix. In oFAPEC transfected with specific Rac1 small interfering RNA (siRNA, 40 nm), VEGF-induced intracellular superoxide formation was completely abrogated in association with a 78% reduction of endogenous Rac1. In oFPAE cells transfected with the specific Rac1 siRNA, but not with transfection reagent alone or scrambled control siRNA, VEGF-induced cell proliferation, migration, and tube-like structure formation were dramatically inhibited. Pretreatment of an NADPH oxidase inhibitor apocynin also abrogates the VEGF-stimulated intracellular superoxide production and DNA synthesis in oFPAECs. Taken together, our results demonstrated that a Rac1/Nox2-based NADPH oxidase system is present in placental endothelial cells. This NADPH oxidase system appears to generate the second messenger superoxide that plays a critical role in the signaling control of the VEGF-induced placental angiogenesis.
Collapse
Affiliation(s)
- Su-min Li
- Department of Reproductive Medicine, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
29
|
Liao WX, Feng L, Zheng J, Chen DB. Deciphering mechanisms controlling placental artery endothelial cell migration stimulated by vascular endothelial growth factor. Endocrinology 2010; 151:3432-44. [PMID: 20463056 PMCID: PMC2903938 DOI: 10.1210/en.2009-1305] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vascular endothelial growth factor (VEGF) stimulated fetoplacental artery endothelial (oFPAE) cell migration and activated multiple signaling pathways including ERK2/1, p38MAPK, Jun N-terminal kinase (JNK1/2), v-Akt murine thymoma viral oncogene homolog 1 (Akt1), and c-Src in oFPAE cells. VEGF-induced cell migration was blocked by specific kinase inhibitors of JNK1/2 (SP600125), c-Src (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine), and phosphatidylinositol 3-kinase/Akt (wortmannin) but not ERK2/1 (U0126) and p38MAPK (SB203580). VEGF-induced cell migration was associated with dynamic actin reorganization and focal adhesion as evidenced by increased stress fiber formation and phosphorylation of cofilin-1 and focal adhesion kinase (FAK) and paxillin. Inhibition of JNK1/2, c-Src, and phosphatidylinositol 3-kinase/Akt suppressed VEGF-induced stress fiber formation and cofilin-1 phosphorylation. c-Src inhibition suppressed VEGF-induced phosphorylation of focal adhesion kinase, paxillin, and focal adhesion. VEGF-induced cell migration requires endogenous nitric oxide (NO) as: 1) VEGF-stimulated phosphorylation of endothelial NO synthase (eNOS) via activation of Akt, JNK1/2, and Src; 2) a NO donor diethylenetriamine-NO-stimulated cell migration; and 3) NO synthase inhibition blocked VEGF-induced cell migration. Targeted down-regulation and overexpression of caveolin-1 both inhibited VEGF-induced cell migration. Caveolin-1 down-regulation suppressed VEGF-stimulated phosphorylation of Akt, JNK, eNOS, c-Src, and FAK; however, basal activities of c-Src and FAK were elevated in parallel with increased stress fiber formation and focal adhesion. Caveolin-1 overexpression also inhibited VEGF-induced phosphorylation of Akt, JNK, c-Src, FAK, and eNOS. Thus, VEGF-induced placental endothelial cell migration requires activation of complex pathways that are paradoxically regulated by caveolin-1.
Collapse
Affiliation(s)
- Wu-xiang Liao
- Department of Obstetrics and Gynecology, University of California-Irvine, Orange, CA 92673, USA
| | | | | | | |
Collapse
|
30
|
Davis CW, Hawkins BJ, Ramasamy S, Irrinki KM, Cameron BA, Islam K, Daswani VP, Doonan PJ, Manevich Y, Madesh M. Nitration of the mitochondrial complex I subunit NDUFB8 elicits RIP1- and RIP3-mediated necrosis. Free Radic Biol Med 2010; 48:306-17. [PMID: 19897030 PMCID: PMC2818448 DOI: 10.1016/j.freeradbiomed.2009.11.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 10/15/2009] [Accepted: 11/03/2009] [Indexed: 11/18/2022]
Abstract
Nitric oxide (NO) and other reactive nitrogen species target multiple sites in the mitochondria to influence cellular bioenergetics and survival. Kinetic imaging studies revealed that NO from either activated macrophages or donor compounds rapidly diffuses to the mitochondria, causing a dose-dependent progressive increase in NO-dependent DAF fluorescence, which corresponded to mitochondrial membrane potential loss and initiated alterations in cellular bioenergetics that ultimately led to necrotic cell death. Cellular dysfunction is mediated by an elevated 3-nitrotyrosine signature of the mitochondrial complex I subunit NDUFB8, which is vital for normal mitochondrial function as evidenced by selective knockdown via siRNA. Overexpression of mitochondrial superoxide dismutase substantially decreased NDUFB8 nitration and restored mitochondrial homeostasis. Further, treatment of cells with either necrostatin-1 or siRNA knockdown of RIP1 and RIP3 prevented NO-mediated necrosis. This work demonstrates that the interaction between NO and mitochondrially derived superoxide alters mitochondrial bioenergetics and cell function, thus providing a molecular mechanism for reactive oxygen and nitrogen species-mediated alterations in mitochondrial homeostasis.
Collapse
Affiliation(s)
- Christiana W. Davis
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Brian J. Hawkins
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Subbiah Ramasamy
- Department of Biochemistry, Temple University, Philadelphia, PA 19140
| | - Krishna M. Irrinki
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Biochemistry, Temple University, Philadelphia, PA 19140
| | - Bruce A. Cameron
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Defence Research and Development Canada, Toronto, Ontario, Canada
- Bamfield Marine Sciences Centre, Bamfield, British Columbia, V0R 1B0, Canada
| | - Khalid Islam
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Varsha P. Daswani
- Department of Biochemistry, Temple University, Philadelphia, PA 19140
| | - Patrick J. Doonan
- Department of Biochemistry, Temple University, Philadelphia, PA 19140
| | - Yefim Manevich
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Muniswamy Madesh
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Biochemistry, Temple University, Philadelphia, PA 19140
| |
Collapse
|
31
|
Wang K, Jiang YZ, Chen DB, Zheng J. Hypoxia enhances FGF2- and VEGF-stimulated human placental artery endothelial cell proliferation: roles of MEK1/2/ERK1/2 and PI3K/AKT1 pathways. Placenta 2009; 30:1045-51. [PMID: 19892399 DOI: 10.1016/j.placenta.2009.10.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 10/13/2009] [Accepted: 10/15/2009] [Indexed: 12/22/2022]
Abstract
Placental development occurs under a low oxygen (2-8% O(2)) environment, which is critical for placental development and angiogenesis. In this study, we examined if hypoxia affected fibroblast growth factor-2 (FGF2)- and vascular endothelial growth factor (VEGF)-stimulated cell proliferation via the mitogen-activated protein kinase kinase 1/2 (MEK1/2)/extracellular signal-regulated kinases 1/2 (ERK1/2) and phosphatidylinositol-3 kinase (PI3K)/v-akt murine thymomaviral oncogene homologue (AKT1) pathways in human placental artery endothelial (HPAE) cells. We observed that under normoxia (approximately 20% O(2)), FGF2 and VEGF dose-dependently stimulated cell proliferation. Hypoxia (3% O(2)) significantly promoted FGF2- and VEGF-stimulated cell proliferation as compared to normoxia. Under both normoxia and hypoxia, FGF2 rapidly induced ERK1/2 and AKT1 phosphorylation, while VEGF-induced ERK1/2, but not AKT1 phosphorylation. However, hypoxia did not significantly alter FGF2- and VEGF-induced ERK1/2 and AKT1 phosphorylation as compared to normoxia. PD98059 (a MEK1/2 inhibitor) at 20microM and LY294002 (a PI3K inhibitor) at 5microM attenuated FGF2- and VEGF-induced phosphorylation of ERK1/2 and AKT1, respectively. PD98059, even at doses that drastically inhibited FGF2-induced ERK1/2 phosphorylation (20microM) and caused cell loss (40microM), did not affect FGF2-stimulated cell proliferation, which was confirmed by U0126 (another potent MEK1/2 inhibitor). PD98059, however, dose-dependently inhibited VEGF-stimulated cell proliferation. Conversely, LY294002 dose-dependently inhibited FGF2-, but not VEGF-stimulated cell proliferation. These data suggest that in the MEK1/2/ERK1/2 and PI3K/AKT1 pathways differentially mediate FGF2- and VEGF-stimulated HPAE cell proliferation. These results also indicate that hypoxia promotes FGF2- and VEGF-stimulated cell proliferation without further activation of the PI3K/AKT1 and MEK1/2/ERK1/2, respectively.
Collapse
Affiliation(s)
- K Wang
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin, Madison, WI 53715, USA
| | | | | | | |
Collapse
|
32
|
Fujinaga H, Baker CD, Ryan SL, Markham NE, Seedorf GJ, Balasubramaniam V, Abman SH. Hyperoxia disrupts vascular endothelial growth factor-nitric oxide signaling and decreases growth of endothelial colony-forming cells from preterm infants. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1160-9. [PMID: 19734318 DOI: 10.1152/ajplung.00234.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Exposure of preterm infants to hyperoxia impairs vascular growth, contributing to the development of bronchopulmonary dysplasia and retinopathy of prematurity. Disruption of vascular endothelial growth factor (VEGF)-nitric oxide (NO) signaling impairs vascular growth. Endothelial progenitor cells (EPCs) may play an important role in vascular growth. Endothelial colony-forming cells (ECFCs), a type of EPC, from human preterm cord blood are more susceptible to hyperoxia-induced growth impairment than term ECFCs. Therefore, we hypothesized that hyperoxia disrupts VEGF-NO signaling and impairs growth in preterm ECFCs and that exogenous VEGF or NO preserves growth in hyperoxia. Growth kinetics of preterm cord blood-derived ECFCs (gestational ages, 27-34 wk) were assessed in room air (RA) and hyperoxia (40-50% oxygen) with or without VEGF, NO, or N(omega)-nitro-l-arginine. VEGF, VEGF receptor-2 (VEGFR-2), and endothelial NO synthase (eNOS) protein expression and NO production were compared. Compared with RA controls, hyperoxia significantly decreased growth, VEGFR-2 and eNOS expression, and NO production. VEGF treatment restored growth in hyperoxia to values measured in RA controls and significantly increased eNOS expression in hyperoxia. NO treatment also increased growth in hyperoxia. N(omega)-nitro-l-arginine treatment inhibited VEGF-augmented growth in RA and hyperoxia. We conclude that hyperoxia decreases growth and disrupts VEGF-NO signaling in human preterm ECFCs. VEGF treatment restores growth in hyperoxia by increasing NO production. NO treatment also increases growth during hyperoxia. Exogenous VEGF or NO may protect preterm ECFCs from the adverse effects of hyperoxia and preservation of ECFC function may improve outcomes of preterm infants.
Collapse
Affiliation(s)
- Hideshi Fujinaga
- Pediatric Heart Lung Center. Univ. of Colorado Denver School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Onumah OE, Jules GE, Zhao Y, Zhou L, Yang H, Guo Z. Overexpression of catalase delays G0/G1- to S-phase transition during cell cycle progression in mouse aortic endothelial cells. Free Radic Biol Med 2009; 46:1658-67. [PMID: 19341793 PMCID: PMC2713001 DOI: 10.1016/j.freeradbiomed.2009.03.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 03/23/2009] [Accepted: 03/25/2009] [Indexed: 01/26/2023]
Abstract
Although it is understood that hydrogen peroxide (H(2)O(2)) promotes cellular proliferation, little is known about its role in endothelial cell cycle progression. To assess the regulatory role of endogenously produced H(2)O(2) in cell cycle progression, we studied the cell cycle progression in mouse aortic endothelial cells (MAECs) obtained from mice overexpressing a human catalase transgene (hCatTg), which destroys H(2)O(2). The hCatTg MAECs displayed a prolonged doubling time compared to wild-type controls (44.0 +/- 4.7 h versus 28.6 +/- 0.8 h, p<0.05), consistent with a diminished growth rate and H(2)O(2) release. Incubation with aminotriazole, a catalase inhibitor, prevented the observed diminished growth rate in hCatTg MAECs. Inhibition of catalase activity with aminotriazole abrogated catalase overexpression-induced antiproliferative action. Flow cytometry analysis indicated that the prolonged doubling time was principally due to an extended G(0)/G(1) phase in hCatTg MAECs compared to the wild-type cells (25.0 +/- 0.9 h versus 15.9 +/- 1.4 h, p< 0.05). The hCatTg MAECs also exhibited decreased activities of the cyclin-dependent kinase (Cdk) complexes responsible for G(0)/G(1)- to S-phase transition in the cell cycle, including the cyclin D-Cdk4 and cyclin E-Cdk2 complexes. Moreover, the reduction in cyclin-Cdk activities in hCatTg MAECs was accompanied by increased protein levels of two Cdk inhibitors, p21 and p27, which inhibit the Cdk activity required for the G(0)/G(1)- to S-phase transition. Knockdown of p21 and/or p27 attenuated the antiproliferative effect of catalase overexpression in MAECs. These results, together with the fact that catalase is an H(2)O(2) scavenger, suggest that endogenously produced H(2)O(2) mediates MAEC proliferation by fostering the transition from G(0)/G(1) to S phase.
Collapse
Affiliation(s)
- Ogbeyalu E. Onumah
- Department of Cancer Biology, Meharry Medical College, Nashville, TN 37208
| | - George E. Jules
- Department of Cardiovascular Biology, Meharry Medical College, Nashville, TN 37208
| | - Yanfeng Zhao
- Department of Cardiovascular Biology, Meharry Medical College, Nashville, TN 37208
| | - LiChun Zhou
- Department of Cardiovascular Biology, Meharry Medical College, Nashville, TN 37208
| | - Hong Yang
- Department of Cardiovascular Biology, Meharry Medical College, Nashville, TN 37208
| | - ZhongMao Guo
- Department of Cardiovascular Biology, Meharry Medical College, Nashville, TN 37208
| |
Collapse
|
34
|
Liao WX, Feng L, Zhang H, Zheng J, Moore TR, Chen DB. Compartmentalizing VEGF-induced ERK2/1 signaling in placental artery endothelial cell caveolae: a paradoxical role of caveolin-1 in placental angiogenesis in vitro. Mol Endocrinol 2009; 23:1428-44. [PMID: 19477952 DOI: 10.1210/me.2008-0475] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
On vascular endothelial growth factor (VEGF) stimulation, both VEGF R1 and R2 receptors were phosphorylated in ovine fetoplacental artery endothelial (oFPAE) cells. Treatment with VEGF stimulated both time- and dose-dependent activation of ERK2/1 in oFPAE cells. VEGF-induced ERK2/1 activation was mediated by VEGFR2, but not VEGFR1, and was linked to intracellular calcium, protein kinase C, and Raf-1. VEGF stimulated oFPAE cell proliferation, migration, and tube formation in vitro. Blockade of ERK2/1 pathway attenuated VEGF-induced cell proliferation and tube formation but failed to inhibit migration in oFPAE cells. Disruption of caveolae by cholesterol depletion with methyl-beta-cyclodextrin or by down-regulation of its structural protein caveolin-1 blunted VEGF-induced ERK2/1 activation, proliferation, and tube formation in oFPAE cells, indicating an essential role of integral caveolae in these VEGF-induced responses. Adenoviral overexpression of caveolin-1 and addition of a caveolin scaffolding domain peptide also inhibited VEGF-stimulated ERK2/1 activation, cell proliferation, and tube formation in oFPAE cells. Furthermore, molecules comprising the ERK2/1 signaling module, including VEGFR2, protein kinase Calpha, Raf-1, MAPK kinase 1/2, and ERK2/1, resided with caveolin-1 in caveolae. VEGF transiently stimulated ERK2/1 activation in the caveolae similarly as in intact cells. Caveolae disruption greatly diminished ERK2/1 activation by VEGF in oFPAE cell caveolae. We conclude that caveolae function as a platform for compartmentalizing the VEGF-induced ERK2/1 signaling module. Caveolin-1 and caveolae play a paradoxical role in regulating VEGF-induced ERK2/1 activation and in vitro angiogenesis as evidenced by the similar inhibitory effects of down-regulation and overexpression of caveolin-1 and disruption of caveolae in oFPAE cells.
Collapse
Affiliation(s)
- Wu-Xiang Liao
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | |
Collapse
|
35
|
Down-regulation of growth signaling pathways linked to a reduced cotyledonary vascularity in placentomes of over-nourished, obese pregnant ewes. Placenta 2009; 30:405-10. [PMID: 19268361 DOI: 10.1016/j.placenta.2009.02.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 02/04/2009] [Accepted: 02/07/2009] [Indexed: 12/25/2022]
Abstract
Both protein kinase B (Akt) and extracellular signal-regulated kinase 1/2 (ERK1/2) are down-stream components of the insulin/insulin like growth factor-1 (IGF-1) signaling pathway. AMP-activated protein kinase (AMPK) is known to sensitize cells to insulin/IGF-1 signaling. The objective of this study was to assess the activity of AMPK and its role in the observed down-regulation of insulin/IGF-1 signaling in cotyledonary (COT) arteries supplying the placental component of the ewe placentome. Nonpregnant ewes were randomly assigned to a control (C, 100% of NRC recommendations) or obesogenic (OB, 150% of NRC) diet from 60 days before conception until necropsy on day 75 of gestation (n=5/group) or until lambing (n=5/group). At necropsy on day 75 of gestation, the smallest terminal arteries that entered the COT tissues (0.5-1.0 mm in diameter) were collected for analyses. Fetal weights were approximately 20% greater (P<0.05) on OB than C ewes, but birth weights of lambs were similar across dietary groups. Fetal plasma concentrations of glucose, insulin and IGF-1 were higher (P<0.05) in the blood of fetuses from OB than C ewes. Total AMPK and phosphorylated AMPK at Thr 172 (the active form) were reduced (P<0.05) by 19.7+/-8.4% and 25.9+/-7.7%, respectively in the COT arterial tissues of OB ewes. Total acetyl-CoA carboxylase (ACC), a down-stream target of AMPK, and its phosphorylated form were also reduced (P<0.05) by 32.9+/-9.2% and 45.4+/-14.6%, respectively. The phosphorylation of IRS-1 at Ser 789, a site phosphorylated by AMPK, was 24.5+/-9.0% lower (P<0.05) in COT arteries of OB than C ewes. No alteration in total insulin receptor, total IGF-1 receptor or their phosphorylated forms was observed, down-stream insulin signaling was down-regulated in COT arteries of OB ewes, which may have resulted in the observed decrease in COT vascular development in OB ewes.
Collapse
|
36
|
Abstract
Following an arterial occlusion outward remodeling of pre-existent inter-connecting arterioles occurs by proliferation of vascular smooth muscle and endothelial cells. This is initiated by deformation of the endothelial cells through increased pulsatile fluid shear stress (FSS) caused by the steep pressure gradient between the high pre-occlusive and the very low post-occlusive pressure regions that are interconnected by collateral vessels. Shear stress leads to the activation and expression of all NOS isoforms and NO production, followed by endothelial VEGF secretion, which induces MCP-1 synthesis in endothelium and in the smooth muscle of the media. This leads to attraction and activation of monocytes and T-cells into the adventitial space (peripheral collateral vessels) or attachment of these cells to the endothelium (coronary collaterals). Mononuclear cells produce proteases and growth factors to digest the extra-cellular scaffold and allow motility and provide space for the new cells. They also produce NO from iNOS, which is essential for arteriogenesis. The bulk of new tissue production is carried by the smooth muscles of the media, which transform their phenotype from a contractile into a synthetic and proliferative one. Important roles are played by actin binding proteins like ABRA, cofilin, and thymosin beta 4 which determine actin polymerization and maturation. Integrins and connexins are markedly up-regulated. A key role in this concerted action which leads to a 2-to-20 fold increase in vascular diameter, depending on species size (mouse versus human) are the transcription factors AP-1, egr-1, carp, ets, by the Rho pathway and by the Mitogen Activated Kinases ERK-1 and -2. In spite of the enormous increase in tissue mass (up to 50-fold) the degree of functional restoration of blood flow capacity is incomplete and ends at 30% of maximal conductance (coronary) and 40% in the vascular periphery. The process of arteriogenesis can be drastically stimulated by increases in FSS (arterio-venous fistulas) and can be completely blocked by inhibition of NO production, by pharmacological blockade of VEGF-A and by the inhibition of the Rho-pathway. Pharmacological stimulation of arteriogenesis, important for the treatment of arterial occlusive diseases, seems feasible with NO donors.
Collapse
|
37
|
Sharif F, Hynes SO, Cooney R, Howard L, McMahon J, Daly K, Crowley J, Barry F, O'Brien T. Gene-eluting Stents: Adenovirus-mediated Delivery of eNOS to the Blood Vessel Wall Accelerates Re-endothelialization and Inhibits Restenosis. Mol Ther 2008; 16:1674-80. [DOI: 10.1038/mt.2008.165] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
38
|
Wang K, Song Y, Chen DB, Zheng J. Protein phosphatase 3 differentially modulates vascular endothelial growth factor- and fibroblast growth factor 2-stimulated cell proliferation and signaling in ovine fetoplacental artery endothelial cells. Biol Reprod 2008; 79:704-10. [PMID: 18509162 DOI: 10.1095/biolreprod.108.068957] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A critical process for vascular endothelial growth factor (VEGF)- and fibroblast growth factor 2 (FGF2)-regulated cellular function is reversible protein phosphorylation, which is tightly controlled by a balance of protein kinases and phosphatases. We have reported that in ovine fetoplacental artery endothelial (OFPAE) cells, VEGF and FGF2 stimulate cell proliferation in part via activation of mitogen-activated protein kinase kinase 1/2 (MAP2K1/2)/mitogen-activated protein kinase 3/1 (MAPK3/1) and phosphoinositide 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homolog 1 (AKT1) pathways. In the present study, we examined if protein phosphatase 3 (PPP3) mediated VEGF- and FGF2-stimulated OFPAE cell proliferation via modulating activation of MAPK3/1 and AKT1. Small interfering RNA (siRNA) targeting human PPP3 catalytic subunit alpha (PPP3CA) was used to suppress PPP3CA protein expression in OFPAE cells. Compared with the scrambled siRNA, PPP3CA siRNA decreased PPP3CA protein levels by approximately 97% without altering protein levels of protein phosphatase 2 catalytic subunit alpha, total MAPK3/1, total AKT1, or glyceraldehyde-3-phosphate dehydrogenase. Knockdown of PPP3CA protein expression enhanced VEGF-stimulated, but not FGF2-stimulated, cell proliferation. Knockdown of PPP3CA protein expression did not significantly affect VEGF-induced MAPK3/1 and AKT1 phosphorylation but attenuated FGF2-induced MAPK3/1 and AKT1 phosphorylation. Thus, to our knowledge, the present study is the first to demonstrate successful knockdown of PPP3CA protein expression in any cell model using a single pair of double-strained siRNA. Moreover, specific knockdown of PPP3CA protein expression enhances VEGF-stimulated, but not FGF2-stimulated, OFPAE cell proliferation and attenuates FGF2-induced, but not VEGF-induced, MAPK3/1 and AKT1 activation. Thus, PPP3CA differentially modulates the VEGF- and FGF2-stimulated cell proliferation and signaling cascades in OFPAE cells. These data also suggest that signaling molecules other than MAPK3/1 and AKT1 play an important role in VEGF- and FGF2-stimulated cell proliferation after knockdown of PPP3CA in OFPAE cells.
Collapse
Affiliation(s)
- Kai Wang
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin 53715, USA
| | | | | | | |
Collapse
|
39
|
Zhu MJ, Du M, Hess BW, Nathanielsz PW, Ford SP. Periconceptional Nutrient Restriction in the Ewe Alters MAPK/ERK1/2 and PI3K/Akt Growth Signaling Pathways and Vascularity in the Placentome. Placenta 2007; 28:1192-9. [PMID: 17586041 DOI: 10.1016/j.placenta.2007.05.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 04/20/2007] [Accepted: 05/03/2007] [Indexed: 01/02/2023]
Abstract
This study evaluated the role of MAPK/ERK1/2 and/or PI3K/Akt signaling pathways in modulating ovine placentomal vascularity in response to periconceptional maternal nutrient restriction. Ewes were randomly assigned to be nutrient restricted (NR, 50% NRC recommendation, N=7) or control fed (CF, 100% NRC recommendation, N=7) from 60 +/- 2 days before to 30 days after conception (day 0). From day 31 of gestation, all ewes (CF and NR) were fed the control diet until necropsy on day 78. On day 78 of gestation, NR ewes exhibited greater vascularity in both caruncular (CAR) and cotyledon (COT) tissues than CF ewes. Akt or ERK1/2 content in CAR and COT arterial tissue did not differ across dietary treatment. The activated forms, phosphorylated Akt and phosphorylated ERK1/2, were significantly increased in COT but not CAR arterial tissues of NR ewes compared to those of CF ewes (P<0.05). For both CF and NR ewes, phosphorylated Akt and phosphorylated ERK1/2 content in COT are higher (P<0.05) than those in CAR arterial tissues. Immunohistochemical staining revealed cytoplasmic and nuclear localization of Akt, phosphorylated Akt, ERK1/2 and phosphorylated ERK1/2, with phosphorylated Akt and phosphorylated-ERK1/2 specifically localized in trophoblast cells, while binucleate cells remained unstained. In placentomal blood vessels, Akt, phosphorylated Akt, ERK1/2 and phosphorylated ERK1/2 were localized to both endothelium and smooth muscle cells. These findings demonstrate for the first time that periconceptional NR increases vascular density in both COT than CAR tissues of the ovine placentome, and that the MAPK/ERK1/2 and/or PI3K/Akt signaling pathways are increased in NR COT but not NR CAR arterial tissues.
Collapse
Affiliation(s)
- M J Zhu
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, WY, USA
| | | | | | | | | |
Collapse
|
40
|
Zheng J, Wen Y, Song Y, Wang K, Chen DB, Magness RR. Activation of multiple signaling pathways is critical for fibroblast growth factor 2- and vascular endothelial growth factor-stimulated ovine fetoplacental endothelial cell proliferation. Biol Reprod 2007; 78:143-50. [PMID: 17901071 PMCID: PMC2441762 DOI: 10.1095/biolreprod.107.064477] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Fibroblast growth factor-2 (FGF2) and vascular endothelial growth factor (VEGF) are two key regulators of placental angiogenesis. The potent vasodilator nitric oxide (NO) could also act as a key mediator of FGF2- and VEGF-induced angiogenesis. However, the postreceptor signaling pathways governing these FGF2- and VEGF-induced placental angiogenic responses are poorly understood. In this study, we assessed the role of endogenous NO, mitogen-activated protein kinase 3/1 (MAPK3/1), and v-akt murine thymoma viral oncogene homolog 1 (AKT1) in FGF2- and VEGF-stimulated proliferation of ovine fetoplacental endothelial (OFPAE) cells. Both FGF2 and VEGF time-dependently stimulated (P < 0.05) NO production and activated AKT1. Both FGF2- and VEGF-stimulated cell proliferation was dose-dependently inhibited (P < 0.05) by N(G)-monomethyl-L-arginine (L-NMMA; an NO synthase inhibitor), PD98059 (a selective MAPK3/1 kinase 1 and 2 [MAP2K1/2] inhibitor), or LY294002 (a selective phosphatidylinositol 3 kinase [PI3K] inhibitor) but not by phenyl-4,4,5,5 tetramethylimidazoline-1-oxyl 3-oxide (PTIO, a potent extracellular NO scavenger). At the maximal inhibitory dose without cytotoxicity, PD98059 and LY294002 completely inhibited VEGF-induced cell proliferation but only partially attenuated (P < 0.05) FGF2-induced cell proliferation. PD98059 and LY294002 also inhibited (P < 0.05) FGF2- and VEGF-induced phosphorylation of MAPK3/1 and AKT1, respectively. L-NMMA did not significantly affect FGF2- and VEGF-induced phosphorylation of either MAPK3/1 or AKT1. Thus, in OFPAE cells, both FGF2- and VEGF-stimulated cell proliferation is partly mediated via NO as an intracellular and downstream signal of MAPK3/1 and AKT1 activation. Moreover, activation of both MAP2K1/2/MAPK3/1 and PI3K/AKT1 pathways is critical for FGF2-stimulated cell proliferation, whereas activation of either one pathway is sufficient for mediating the VEGF-induced maximal cell proliferation, indicating that these two kinase pathways differentially mediate the FGF2- and VEGF-stimulated OFPAE cell proliferation.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Perinatal Research Laboratories, PAB1 Meriter Hospital, Madison, WI 53715, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Powell JA, Mousa SA. Neutrophil-activating protein-2- and interleukin-8-mediated angiogenesis. J Cell Biochem 2007; 102:412-20. [PMID: 17352409 DOI: 10.1002/jcb.21302] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In this study, we investigated the anti-angiogenic potential of nitric oxide (NO) donors and anti-integrin compounds against neutrophil-activating protein-2 (NAP-2), interleukin-8 (IL-8), and basic fibroblast growth factor (FGF-2)-induced angiogenesis. In vivo, recombinant human NAP-2 and FGF-2 induce a potent and comparable angiogenic response in the chick embryo chorioallantoic membrane (CAM). We demonstrate that NO donors and anti-integrin agents are capable of abrogating either NAP-2- or FGF-2-induced angiogenesis in the CAM model. The NO donor, S-nitroso N-acetyl penicillamine (SNAP), blocked either NAP-2- or FGF-2-mediated angiogenesis in the CAM. Similarly, angiogenesis stimulated with NAP-2 or FGF-2 was blocked by antagonist of the alphavbeta3 integrin in the CAM model. However, the inhibition of NAP-2 and IL-8 by the anti-integrin compound is significantly less than the inhibition observed with FGF-2 as the angiogenic stimulus. Similarly, the ability of these mechanisms to also inhibit endothelial cell differentiation was demonstrated. Taken together, these data illustrate the involvement of multiple pathways in the mechanisms of action for the alpha-chemokine- and cytokine-mediated angiogenesis. These approaches may be a useful tool for the inhibition of angiogenesis associated with human tumor growth or with neovascular, ocular, and inflammatory diseases where chemokines and cytokines are involved.
Collapse
Affiliation(s)
- John A Powell
- University of Pennsylvania, Medical School, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
42
|
Wu G, Bazer FW, Wallace JM, Spencer TE. BOARD-INVITED REVIEW: Intrauterine growth retardation: Implications for the animal sciences1. J Anim Sci 2006; 84:2316-37. [PMID: 16908634 DOI: 10.2527/jas.2006-156] [Citation(s) in RCA: 785] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intrauterine growth retardation (IUGR), defined as impaired growth and development of the mammalian embryo/fetus or its organs during pregnancy, is a major concern in domestic animal production. Fetal growth restriction reduces neonatal survival, has a permanent stunting effect on postnatal growth and the efficiency of feed/forage utilization in offspring, negatively affects whole body composition and meat quality, and impairs long-term health and athletic performance. Knowledge of the underlying mechanisms has important implications for the prevention of IUGR and is crucial for enhancing the efficiency of livestock production and animal health. Fetal growth within the uterus is a complex biological event influenced by genetic, epigenetic, and environmental factors, as well as maternal maturity. These factors impact on the size and functional capacity of the placenta, uteroplacental blood flows, transfer of nutrients and oxygen from mother to fetus, conceptus nutrient availability, the endocrine milieu, and metabolic pathways. Alterations in fetal nutrition and endocrine status may result in developmental adaptations that permanently change the structure, physiology, metabolism, and postnatal growth of the offspring. Impaired placental syntheses of nitric oxide (a major vasodilator and angiogenic factor) and polyamines (key regulators of DNA and protein synthesis) may provide a unified explanation for the etiology of IUGR in response to maternal undernutrition and overnutrition. There is growing evidence that maternal nutritional status can alter the epigenetic state (stable alterations of gene expression through DNA methylation and histone modifications) of the fetal genome. This may provide a molecular mechanism for the role of maternal nutrition on fetal programming and genomic imprinting. Innovative interdisciplinary research in the areas of nutrition, reproductive physiology, and vascular biology will play an important role in designing the next generation of nutrient-balanced gestation diets and developing new tools for livestock management that will enhance the efficiency of animal production and improve animal well being.
Collapse
Affiliation(s)
- G Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA.
| | | | | | | |
Collapse
|