1
|
Mancinetti F, Labarile F, Bastiani P, Scamosci M, Alunno M, Cecchetti R, Mecocci P, Boccardi V. A novel sex-specific association between insulin resistance and depressive symptoms in older adults: The potential mediating role of Vascular Endothelial Growth Factor. J Affect Disord 2025; 382:194-200. [PMID: 40274107 DOI: 10.1016/j.jad.2025.04.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/08/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Insulin resistance (IR) and depression are increasingly recognized as interconnected conditions in aging, potentially linked through chronic low-grade inflammation ("inflammaging"). The triglyceride-glucose (TyG) index has emerged as a validated surrogate marker of IR, yet its relationship with inflammatory biomarkers and depressive symptoms in older adults remains underexplored. OBJECTIVE This study investigated the association between the TyG index, depressive symptoms, and circulating inflammatory molecules in cognitively healthy older adults, with emphasis on sex-specific differences and the potential mediating role of vascular endothelial growth factor (VEGF). METHODS In this retrospective study, 118 non-diabetic older adults (mean age 74.1 years; 52.5 % men) with preserved cognition (MMSE ≥27) were assessed. Depressive symptoms were measured using the Geriatric Depression Scale (GDS), while cytokines and growth factors were quantified via multiplex immunoassay. The TyG index was calculated from fasting glucose and triglyceride levels. Multiple linear regression models controlled for age, sex, BMI, and medication use. RESULTS Depressive symptoms (GDS ≥5) were present in 31.35 % of participants. Those with depression had lower levels of anti-inflammatory cytokines and elevated levels of VEGF and MCP-1. TyG index correlated with both GDS scores (r = 0.239, p = 0.017) and VEGF (r = 0.271, p = 0.005), with significant associations observed only in women. VEGF emerged as a key mediator in the TyG-depression link in women (R2 = 0.425). CONCLUSIONS These findings suggest a sex-specific metabolic-inflammatory signature in late-life depression. VEGF may serve as a mechanistic link between IR and depressive symptoms in older women, supporting sex-tailored interventions.
Collapse
Affiliation(s)
- Francesca Mancinetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Flavia Labarile
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Patrizia Bastiani
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Michela Scamosci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Martina Alunno
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Roberta Cecchetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy; Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy.
| |
Collapse
|
2
|
Islam MT, Cai J, Allen S, Moreno DG, Bloom SI, Bramwell RC, Mitton J, Horn AG, Zhu W, Donato AJ, Holland WL, Lesniewski LA. Endothelial-Specific Reduction in Arf6 Impairs Insulin-Stimulated Vasodilation and Skeletal Muscle Blood Flow Resulting in Systemic Insulin Resistance in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1101-1113. [PMID: 38545783 PMCID: PMC11042974 DOI: 10.1161/atvbaha.123.319375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 02/27/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Much of what we know about insulin resistance is based on studies from metabolically active tissues such as the liver, adipose tissue, and skeletal muscle. Emerging evidence suggests that the vascular endothelium plays a crucial role in systemic insulin resistance; however, the underlying mechanisms remain incompletely understood. Arf6 (ADP ribosylation factor 6) is a small GTPase that plays a critical role in endothelial cell function. Here, we tested the hypothesis that the deletion of endothelial Arf6 will result in systemic insulin resistance. METHODS We used mouse models of constitutive endothelial cell-specific Arf6 deletion (Arf6f/- Tie2Cre+) and tamoxifen-inducible Arf6 knockout (Arf6f/f Cdh5CreER+). Endothelium-dependent vasodilation was assessed using pressure myography. Metabolic function was assessed using a battery of metabolic assessments including glucose and insulin tolerance tests and hyperinsulinemic-euglycemic clamps. We used a fluorescence microsphere-based technique to measure tissue blood flow. Skeletal muscle capillary density was assessed using intravital microscopy. RESULTS Endothelial Arf6 deletion impaired insulin-stimulated vasodilation in white adipose tissue and skeletal muscle feed arteries. The impairment in vasodilation was primarily due to attenuated insulin-stimulated nitric oxide bioavailability but independent of altered acetylcholine-mediated or sodium nitroprusside-mediated vasodilation. Endothelial cell-specific deletion of Arf6 also resulted in systematic insulin resistance in normal chow-fed mice and glucose intolerance in high-fat diet-fed obese mice. The underlying mechanisms of glucose intolerance were reductions in insulin-stimulated blood flow and glucose uptake in the skeletal muscle and were independent of changes in capillary density or vascular permeability. CONCLUSIONS Results from this study support the conclusion that endothelial Arf6 signaling is essential for maintaining insulin sensitivity. Reduced expression of endothelial Arf6 impairs insulin-mediated vasodilation and results in systemic insulin resistance. These results have therapeutic implications for diseases that are associated with endothelial cell dysfunction and insulin resistance such as diabetes.
Collapse
Affiliation(s)
- Md Torikul Islam
- Department of Nutrition and Integrative Physiology (M.T.I., S.I.B., A.J.D., W.L.H., L.A.L.), The University of Utah, Salt Lake City
| | - Jinjin Cai
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
| | - Shanena Allen
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
| | - Denisse G Moreno
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
| | - Samuel I Bloom
- Department of Nutrition and Integrative Physiology (M.T.I., S.I.B., A.J.D., W.L.H., L.A.L.), The University of Utah, Salt Lake City
| | - R Colton Bramwell
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
| | - Jonathan Mitton
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
| | - Andrew G Horn
- Department of Kinesiology, Kansas State University, Manhattan (A.G.H.)
| | - Weiquan Zhu
- Division of Cardiovascular Medicine, Department of Internal Medicine (W.Z.), The University of Utah, Salt Lake City
- Department of Pathology (W.Z.), The University of Utah, Salt Lake City
- Program of Molecular Medicine (W.Z.), The University of Utah, Salt Lake City
| | - Anthony J Donato
- Department of Nutrition and Integrative Physiology (M.T.I., S.I.B., A.J.D., W.L.H., L.A.L.), The University of Utah, Salt Lake City
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
- Department of Biochemistry (A.J.D.), The University of Utah, Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.J.D., L.A.L.), The University of Utah, Salt Lake City
- Veteran's Affairs Medical Center-Salt Lake City, Geriatric Research and Clinical Center, UT (A.J.D., L.A.L.)
| | - William L Holland
- Department of Nutrition and Integrative Physiology (M.T.I., S.I.B., A.J.D., W.L.H., L.A.L.), The University of Utah, Salt Lake City
| | - Lisa A Lesniewski
- Department of Nutrition and Integrative Physiology (M.T.I., S.I.B., A.J.D., W.L.H., L.A.L.), The University of Utah, Salt Lake City
- Division of Geriatrics, Department of Internal Medicine (J.C., S.A., D.G.M., R.C.B., J.M., A.J.D., L.A.L.), The University of Utah, Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.J.D., L.A.L.), The University of Utah, Salt Lake City
- Veteran's Affairs Medical Center-Salt Lake City, Geriatric Research and Clinical Center, UT (A.J.D., L.A.L.)
| |
Collapse
|
3
|
Nguyen TT, Corvera S. Adipose tissue as a linchpin of organismal ageing. Nat Metab 2024; 6:793-807. [PMID: 38783156 PMCID: PMC11238912 DOI: 10.1038/s42255-024-01046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Ageing is a conserved biological process, modulated by intrinsic and extrinsic factors, that leads to changes in life expectancy. In humans, ageing is characterized by greatly increased prevalence of cardiometabolic disease, type 2 diabetes and disorders associated with impaired immune surveillance. Adipose tissue displays species-conserved, temporal changes with ageing, including redistribution from peripheral to central depots, loss of thermogenic capacity and expansion within the bone marrow. Adipose tissue is localized to discrete depots, and also diffusely distributed within multiple organs and tissues in direct proximity to specialized cells. Thus, through their potent endocrine properties, adipocytes are capable of modulating tissue and organ function throughout the body. In addition to adipocytes, multipotent progenitor/stem cells in adipose tissue play a crucial role in maintenance and repair of tissues throughout the lifetime. Adipose tissue may therefore be a central driver for organismal ageing and age-associated diseases. Here we review the features of adipose tissue during ageing, and discuss potential mechanisms by which these changes affect whole-body metabolism, immunity and longevity. We also explore the potential of adipose tissue-targeted therapies to ameliorate age-associated disease burdens.
Collapse
Affiliation(s)
- Tammy T Nguyen
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center, Worcester, MA, USA
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - Silvia Corvera
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA.
- Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, USA.
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Cao Y, Langer R, Ferrara N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat Rev Drug Discov 2023; 22:476-495. [PMID: 37041221 DOI: 10.1038/s41573-023-00671-z] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 04/13/2023]
Abstract
Angiogenesis is an essential process in normal development and in adult physiology, but can be disrupted in numerous diseases. The concept of targeting angiogenesis for treating diseases was proposed more than 50 years ago, and the first two drugs targeting vascular endothelial growth factor (VEGF), bevacizumab and pegaptanib, were approved in 2004 for the treatment of cancer and neovascular ophthalmic diseases, respectively. Since then, nearly 20 years of clinical experience with anti-angiogenic drugs (AADs) have demonstrated the importance of this therapeutic modality for these disorders. However, there is a need to improve clinical outcomes by enhancing therapeutic efficacy, overcoming drug resistance, defining surrogate markers, combining with other drugs and developing the next generation of therapeutics. In this Review, we examine emerging new targets, the development of new drugs and challenging issues such as the mode of action of AADs and elucidating mechanisms underlying clinical benefits; we also discuss possible future directions of the field.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
Islam MT, Cai J, Allen S, Moreno DG, Bloom SI, Bramwell RC, Mitton J, Horn AG, Zhu W, Donato AJ, Holland WL, Lesniewski LA. Endothelial specific reduction in Arf6 impairs insulin-stimulated vasodilation and skeletal muscle blood flow resulting in systemic insulin resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539173. [PMID: 37205339 PMCID: PMC10187242 DOI: 10.1101/2023.05.02.539173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Much of what we know about insulin resistance is based on studies from metabolically active tissues such as liver, adipose tissue, and skeletal muscle. Emerging evidence suggests that the vascular endothelium plays a crucial role in systemic insulin resistance, however, the underlying mechanisms remain incompletely understood. ADP ribosylation factor 6 (Arf6) is a small GTPase that plays a critical role in endothelial cell (EC) function. Here, we tested the hypothesis that the deletion of endothelial Arf6 will result in systemic insulin resistance. Methods We used mouse models of constitutive EC-specific Arf6 deletion (Arf6 f/- Tie2Cre) and tamoxifen inducible Arf6 knockout (Arf6 f/f Cdh5Cre). Endothelium-dependent vasodilation was assessed using pressure myography. Metabolic function was assessed using a battery of metabolic assessments including glucose- and insulin-tolerance tests and hyperinsulinemic-euglycemic clamps. A fluorescence microsphere-based technique was used to measure tissue blood flow. Intravital microscopy was used to assess skeletal muscle capillary density. Results Endothelial Arf6 deletion impaired insulin-stimulated vasodilation in white adipose tissue (WAT) and skeletal muscle feed arteries. The impairment in vasodilation was primarily due to attenuated insulin-stimulated nitric oxide (NO) bioavailability but independent of altered acetylcholine- or sodium nitroprusside-mediated vasodilation. In vitro Arf6 inhibition resulted in suppressed insulin stimulated phosphorylation of Akt and endothelial NO synthase. Endothelial cell-specific deletion of Arf6 also resulted in systematic insulin resistance in normal chow fed mice and glucose intolerance in high fat diet fed obese mice. The underlying mechanisms of glucose intolerance were reductions in insulin-stimulated blood flow and glucose uptake in the skeletal muscle and were independent of changes in capillary density or vascular permeability. Conclusion Results from this study support the conclusion that endothelial Arf6 signaling is essential for maintaining insulin sensitivity. Reduced expression of endothelial Arf6 impairs insulin-mediated vasodilation and results in systemic insulin resistance. These results have therapeutic implications for diseases that are associated with endothelial cell dysfunction and insulin resistance such as diabetes.
Collapse
|
6
|
Smad4-mediated angiogenesis facilitates the beiging of white adipose tissue in mice. iScience 2023; 26:106272. [PMID: 36915676 PMCID: PMC10005906 DOI: 10.1016/j.isci.2023.106272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 01/12/2023] [Accepted: 02/19/2023] [Indexed: 03/12/2023] Open
Abstract
Beige adipocytes are thermogenic with high expression of uncoupling protein 1 in the white adipose tissue (WAT), accompanied by angiogenesis. Previous studies showed that Smad4 is important for angiogenesis. Here we studied whether endothelial Smad4-mediated angiogenesis is involved in WAT beiging. Inducible knockout of endothelial cell (EC) selective Smad4 (Smad4 iEC-KO) was achieved by using the Smad4 Floxp/floxp and Tie2 CreERT2 mice. Beige fat induction achieved by cold or adrenergic agonist, and angiogenesis were attenuated in WAT of Smad4 iEC-KO mice, with the less proliferation of ECs and adipogenic precursors. RNA sequencing of human ECs showed that Smad4 is involved in angiogenesis-related pathways. Knockdown of SMAD4 attenuated the upregulation of VEGFA, PDGFA, and angiogenesis in vitro. Treatment of human ECs with palmitic acid-induced Smad1/5 phosphorylation and the upregulation of core endothelial genes. Our study shows that endothelial Smad4 is involved in WAT beiging through angiogenesis and the expansion of adipose precursors into beige adipocytes.
Collapse
|
7
|
Zhang Y, Li G, Xiao F, Wang B, Li J, Jia X, Sun Y, Chen H. Relationship between serum fibroblast growth factor 19 and vascular endothelial growth factor and soluble klotho protein in type 1 diabetic children. BMC Pediatr 2023; 23:120. [PMID: 36927328 PMCID: PMC10018886 DOI: 10.1186/s12887-023-03938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Fibroblast growth factor 19 (FGF19) takes part in maintaining the balance of glycolipids and may be involved in complications of type 1 diabetes(T1D) in children. This study aimed at at evaluating the relationship among the levels of serum FGF19 and vascular endothelial growth factor(VEGF)and soluble klotho protein(sklotho) in type 1 diabetic children. METHODS In a cross-section single center study samples were obtained from 96 subjects: 66 T1D and 30 healthy children.Serum FGF19 and VEGF and sklotho concentrations were measured by ELISA. And 66 type 1 diabetes participants were divided into two groups according to T1D duration or three groups according to HbA1c.Furthermore,we compared the serum levels of FGF19 and VEGF and sklotho in different groups. RESULTS The concentration of FGF19 was lower in T1D than in the controls(226.52 ± 20.86pg/mu vs.240.08 ± 23.53 pg/L, p = 0.03),while sklotho was also lower in T1D than in the controls (2448.67 ± 791.92pg/mL vs. 3083.55 ± 1113.47pg/mL, p = 0.011). In contrast, VEGF levels were higher in diabetic patients than in controls (227.95 ± 48.65pg/mL vs. 205.92 ± 28.27 pg/mL, p = 0.016). In T1D, FGF19 and VEGF and sklotho was not correlated with the duration of diabetes. FGF19 and VEGF and sklotho were correlated with HbA1c (r=-0.349, p = 0.004 and r = 0.302, p = 0.014 and r=-0.342, p = 0.005, respectively), but not with blood glucose and lipid. Among subjects in the T1D group, concentrations of FGF19,VEGF and sklotho protein were different between different groups according to the degree of HbA1c(P < 0.005).Furthermore, there was a positive correlation between the serum FGF19 concentration and sklotho levels (r = 0.247,p = 0.045), and a negative correlation between the serum FGF19 concentration and VEGF level(r=-0.335,P = 0.006). CONCLUSIONS The serum FGF19 levels have a close relation with serum VEGF levels and sklotho levels among T1D subjects. FGF19 may be involved in the development of complications in children with type 1 diabetes through interaction with VEGF and sklotho.
Collapse
Affiliation(s)
- Yanjun Zhang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Shandong University, 9677 Jingshi Road, Lixia Area, 250021, Jinan, Shandong, China
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| | - Guimei Li
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Shandong University, 9677 Jingshi Road, Lixia Area, 250021, Jinan, Shandong, China.
- Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, 250021, Jinan, Shandong, China.
| | - Feifei Xiao
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| | - Bang Wang
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| | - Jianchang Li
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| | - Xiuhong Jia
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| | - Yan Sun
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Shandong University, 9677 Jingshi Road, Lixia Area, 250021, Jinan, Shandong, China
- Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, 250021, Jinan, Shandong, China
| | - Hongye Chen
- Department of Pediatrics, Bin Zhou Medical University Hospital, 661 Huangheer Road, 256603, Bin cheng Area, Bin Zhou, Shandong, China
| |
Collapse
|
8
|
Yin T, Chen S, Zeng G, Yuan W, Lu Y, Zhang Y, Huang Q, Xiong X, Xu B, Huang Q. Angiogenesis-Browning Interplay Mediated by Asprosin-Knockout Contributes to Weight Loss in Mice with Obesity. Int J Mol Sci 2022; 23:16166. [PMID: 36555807 PMCID: PMC9783228 DOI: 10.3390/ijms232416166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Asprosin (ASP) is a recently identified adipokine secreted by white adipose tissue (WAT). It plays important roles in the maintenance of glucose homeostasis in the fasting state and in the occurrence and development of obesity. However, there is no report on whether and how ASP would inhibit angiogenesis and fat browning in the mouse adipose microenvironment. Therefore, the study sought to investigate the effects of ASP-knockout on angiogenesis and fat browning, and to identify the interaction between them in the ASP-knockout mouse adipose microenvironment. In the experiments in vivo, the ASP-knockout alleviated the obesity induced by a high fat diet (HFD) and increased the expressions of the browning-related proteins including uncoupling protein 1 (UCP1), PRD1-BF-1-RIZ1 homologus domain-containing protein-16 (PRDM16) and PPAR gamma coactivator 1 (PGC1-α) and the endothelial cell marker (CD31). In the experiments in vitro, treatment with the conditional medium (CM) from ASP-knockout adipocytes (ASP-/--CM) significantly promoted the proliferation, migration and angiogenesis of vascular endothelial cells, and increased the expressions of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/endothelial nitric oxide synthase (eNOS) pathway proteins. In addition, the treatment with CM from endothelial cells (EC-CM) markedly reduced the accumulation of lipid droplets and increased the expressions of the browning-related proteins and the mitochondrial contents. Moreover, the treatment with EC-CM significantly improved the energy metabolism in 3T3-L1 adipocytes. These results highlight that ASP-knockout can promote the browning and angiogenesis of WAT, and the fat browning and angiogenesis can interact in the mouse adipose microenvironment, which contributes to weight loss in the mice with obesity.
Collapse
Affiliation(s)
- Tingting Yin
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Sheng Chen
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Yanli Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Yanan Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Qianqian Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Xiaowei Xiong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Baohua Xu
- Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang 330006, China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
9
|
Cao Y. Blood vessels in fat tissues and vasculature-derived signals in controlling lipid metabolism and metabolic disease. Chin Med J (Engl) 2022; 135:2647-2652. [PMID: 36382988 PMCID: PMC9943976 DOI: 10.1097/cm9.0000000000002406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 65 Stockholm, Sweden
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| |
Collapse
|
10
|
Yang Y, Cao Y. The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol 2022; 86:251-261. [PMID: 35307547 DOI: 10.1016/j.semcancer.2022.03.011] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023]
Abstract
Metastasis is the leading cause of cancer-associated mortality and the underlying mechanisms of cancer metastasis remain elusive. Both blood and lymphatic vasculatures are essential structures for mediating distal metastasis. The vasculature plays multiple functions, including accelerating tumor growth, sustaining the tumor microenvironment, supplying growth and invasive signals, promoting metastasis, and causing cancer-associated systemic disease. VEGF is one of the key angiogenic factors in tumors and participates in the initial stage of tumor development, progression and metastasis. Consequently, VEGF and its receptor-mediated signaling pathways have become one of the most important therapeutic targets for treating various cancers. Today, anti-VEGF-based antiangiogenic drugs (AADs) are widely used in the clinic for treating different types of cancer in human patients. Despite nearly 20-year clinical experience with AADs, the impact of these drugs on cancer metastasis and systemic disease remains largely unknown. In this review article, we focus our discussion on tumor VEGF in cancer metastasis and systemic disease and mechanisms underlying AADs in clinical benefits.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institute, 171 77 Stockholm, Sweden.
| |
Collapse
|
11
|
Yao H, He S. Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review). Mol Med Rep 2021; 24:866. [PMID: 34676881 PMCID: PMC8554381 DOI: 10.3892/mmr.2021.12506] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/15/2021] [Indexed: 01/08/2023] Open
Abstract
Adipocytes are a type of stromal cell found in numerous different tissues that serve an active role in the tumor microenvironment. Cancer-associated adipocytes (CAAs) display a malignant phenotype and are found at the invasive tumor front, which mediates the crosstalk network between adipocytes (the precursor cells that will become cancer-associated adipocytes in the future) and cancer cells. The present review covers the mechanisms of adipocytes in the development of cancer, including metabolic reprogramming, chemotherapy resistance and adipokine regulation. Furthermore, the potential mechanisms involved in the adipocyte-cancer cell cycle in various types of cancer, including breast, ovarian, colon and rectal cancer, are discussed. Deciphering the complex network of CAA-cancer cell crosstalk will provide insights into tumor biology and optimize therapeutic strategies.
Collapse
Affiliation(s)
- Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
12
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Pathophysiology of Physical Inactivity-Dependent Insulin Resistance: A Theoretical Mechanistic Review Emphasizing Clinical Evidence. J Diabetes Res 2021; 2021:7796727. [PMID: 34660812 PMCID: PMC8516544 DOI: 10.1155/2021/7796727] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
The modern lifestyle has a negative impact on health. It is usually accompanied by increased stress levels and lower physical activity, which interferes with body homeostasis. Diabetes mellitus is a relatively common metabolic disorder with increasing prevalence globally, associated with various risk factors, including lower physical activity and a sedentary lifestyle. It has been shown that sedentary behavior increases the risk of insulin resistance, but the intermediate molecular mechanisms are not fully understood. In this mechanistic review, we explore the possible interactions between physical inactivity and insulin resistance to help better understand the pathophysiology of physical inactivity-dependent insulin resistance and finding novel interventions against these deleterious pathways.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, UK
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Sun X, Wu J, Qiang B, Romagnuolo R, Gagliardi M, Keller G, Laflamme MA, Li RK, Nunes SS. Transplanted microvessels improve pluripotent stem cell-derived cardiomyocyte engraftment and cardiac function after infarction in rats. Sci Transl Med 2021; 12:12/562/eaax2992. [PMID: 32967972 DOI: 10.1126/scitranslmed.aax2992] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 05/06/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) offer an unprecedented opportunity to remuscularize infarcted human hearts. However, studies have shown that most hiPSC-CMs do not survive after transplantation into the ischemic myocardial environment, limiting their regenerative potential and clinical application. We established a method to improve hiPSC-CM survival by cotransplanting ready-made microvessels obtained from adipose tissue. Ready-made microvessels promoted a sixfold increase in hiPSC-CM survival and superior functional recovery when compared to hiPSC-CMs transplanted alone or cotransplanted with a suspension of dissociated endothelial cells in infarcted rat hearts. Microvessels showed unprecedented persistence and integration at both early (~80%, week 1) and late (~60%, week 4) time points, resulting in increased vessel density and graft perfusion, and improved hiPSC-CM maturation. These findings provide an approach to cell-based therapies for myocardial infarction, whereby incorporation of ready-made microvessels can improve functional outcomes in cell replacement therapies.
Collapse
Affiliation(s)
- Xuetao Sun
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Jun Wu
- Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Mark Gagliardi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada.,Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5G 2N2, Canada.,Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Heart and Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada.,Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada. .,Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Heart and Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON M5S 3H2, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
14
|
Chen HJ, Li GL, Zhang WX, Fan J, Hu L, Zhang L, Zhang J, Yan YE. Maternal nicotine exposure during pregnancy and lactation induces brown adipose tissue whitening in female offspring. Toxicol Appl Pharmacol 2020; 409:115298. [PMID: 33091441 DOI: 10.1016/j.taap.2020.115298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/11/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023]
Abstract
Maternal nicotine exposure during pregnancy and lactation is associated with obesity in female offspring. Brown adipose tissue (BAT) is related to energy metabolism and obesity. In this study, we explored the mechanism of maternal nicotine exposure on BAT "whitening" in female offspring. Pregnant rats were randomly assigned to nicotine (1.0 mg/kg twice per day, subcutaneous administration) or control groups. The weight, structure, and microvascular density of interscapular BAT (iBAT) and the expression of PGC-1αUCP1 signals, mitochondrial biogenesis-related genes and angiogenesis-related genes were tested in 4- and 26-week-aged female offspring. In vitro, C3H10T1/2 cells were induced to differentiate into mature brown adipocytes, and 0-50 μM nicotine was treated on cells during the differentiation process. Nicotine-exposed females had higher iBAT weight, white-like adipocytes and abnormal mitochondrial structure in iBAT at 26 weeks rather than 4 weeks. The PGC-1αUCP1 signals and brown-like genes were down-regulated at 26 weeks, but the microvascular density and the expression of pro-angiogenic factors reduced more at 4 weeks in the nicotine group. In vitro, 50 μM nicotine significantly decreased the expression of PGC-1αUCP1 signals and angiogenesis-related genes. In conclusion, maternal nicotine exposure during pregnancy and lactation led to the "whitening" of BAT in adult female offspring: nicotine decreased BAT angiogenesis in the early development stage, and then, the impairment of blood vessels programed for the reduction of BAT phenotype through down-regulating the PGC-1αUCP1 signals in adulthood. This impairment of BAT may be a potential mechanism of nicotine-induced obesity in female offspring.
Collapse
Affiliation(s)
- Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jing Zhang
- Center for Animal Experiment/Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
15
|
Thompson JA, Koestler DC. Equivalent change enrichment analysis: assessing equivalent and inverse change in biological pathways between diverse experiments. BMC Genomics 2020; 21:180. [PMID: 32093613 PMCID: PMC7041296 DOI: 10.1186/s12864-020-6589-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background In silico functional genomics have become a driving force in the way we interpret and use gene expression data, enabling researchers to understand which biological pathways are likely to be affected by the treatments or conditions being studied. There are many approaches to functional genomics, but a number of popular methods determine if a set of modified genes has a higher than expected overlap with genes known to function as part of a pathway (functional enrichment testing). Recently, researchers have started to apply such analyses in a new way: to ask if the data they are collecting show similar disruptions to biological functions compared to reference data. Examples include studying whether similar pathways are perturbed in smokers vs. users of e-cigarettes, or whether a new mouse model of schizophrenia is justified, based on its similarity in cytokine expression to a previously published model. However, there is a dearth of robust statistical methods for testing hypotheses related to these questions and most researchers resort to ad hoc approaches. The goal of this work is to develop a statistical approach to identifying gene pathways that are equivalently (or inversely) changed across two experimental conditions. Results We developed Equivalent Change Enrichment Analysis (ECEA). This is a new type of gene enrichment analysis based on a statistic that we call the equivalent change index (ECI). An ECI of 1 represents a gene that was over or under-expressed (compared to control) to the same degree across two experiments. Using this statistic, we present an approach to identifying pathways that are changed in similar or opposing ways across experiments. We compare our approach to current methods on simulated data and show that ECEA is able to recover pathways exhibiting such changes even when they exhibit complex patterns of regulation, which other approaches are unable to do. On biological data, our approach recovered pathways that appear directly connected to the condition being studied. Conclusions ECEA provides a new way to perform gene enrichment analysis that allows researchers to compare their data to existing datasets and determine if a treatment will cause similar or opposing genomic perturbations.
Collapse
|
16
|
Hasan SS, Fischer A. The Endothelium: An Active Regulator of Lipid and Glucose Homeostasis. Trends Cell Biol 2020; 31:37-49. [PMID: 33129632 DOI: 10.1016/j.tcb.2020.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
The vascular endothelium serves as a dynamic barrier that separates blood from interstitia. Endothelial cells (ECs) respond rapidly to changes in the circulation and actively regulate vessel tone, permeability, and platelet functions. ECs also secrete angiocrine factors that dictate the function of adjacent parenchymal cells in an organ-specific manner. Endothelial dysfunction is considered as a hallmark of metabolic diseases. However, there is emerging evidence that ECs modulate the transfer of nutrients and hormones to parenchymal cells in response to alterations in metabolic profile. As such, a causal role for ECs in systemic metabolic dysregulation can be envisaged. This review summarizes recent progress in the understanding of regulated fatty acid, glucose, and insulin transport across the endothelium and discusses its pathophysiological implications.
Collapse
Affiliation(s)
- Sana S Hasan
- Division of Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Fischer
- Division of Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, 69120 Heidelberg, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
17
|
Fang Y, Kaszuba T, Imoukhuede PI. Systems Biology Will Direct Vascular-Targeted Therapy for Obesity. Front Physiol 2020; 11:831. [PMID: 32760294 PMCID: PMC7373796 DOI: 10.3389/fphys.2020.00831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Healthy adipose tissue expansion and metabolism during weight gain require coordinated angiogenesis and lymphangiogenesis. These vascular growth processes rely on the vascular endothelial growth factor (VEGF) family of ligands and receptors (VEGFRs). Several studies have shown that controlling vascular growth by regulating VEGF:VEGFR signaling can be beneficial for treating obesity; however, dysregulated angiogenesis and lymphangiogenesis are associated with several chronic tissue inflammation symptoms, including hypoxia, immune cell accumulation, and fibrosis, leading to obesity-related metabolic disorders. An ideal obesity treatment should minimize adipose tissue expansion and the advent of adverse metabolic consequences, which could be achieved by normalizing VEGF:VEGFR signaling. Toward this goal, a systematic investigation of the interdependency of vascular and metabolic systems in obesity and tools to predict personalized treatment ranges are necessary to improve patient outcomes through vascular-targeted therapies. Systems biology can identify the critical VEGF:VEGFR signaling mechanisms that can be targeted to regress adipose tissue expansion and can predict the metabolic consequences of different vascular-targeted approaches. Establishing a predictive, biologically faithful platform requires appropriate computational models and quantitative tissue-specific data. Here, we discuss the involvement of VEGF:VEGFR signaling in angiogenesis, lymphangiogenesis, adipogenesis, and macrophage specification – key mechanisms that regulate adipose tissue expansion and metabolism. We then provide useful computational approaches for simulating these mechanisms, and detail quantitative techniques for acquiring tissue-specific parameters. Systems biology, through computational models and quantitative data, will enable an accurate representation of obese adipose tissue that can be used to direct the development of vascular-targeted therapies for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yingye Fang
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Tomasz Kaszuba
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - P I Imoukhuede
- Imoukhuede Systems Biology Laboratory, Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
18
|
Chen HJ, Zhang WX, Hu L, Fan J, Zhang L, Yan YE. Maternal nicotine exposure enhances adipose tissue angiogenic activity in offspring: Sex and age differences. Toxicology 2020; 441:152506. [PMID: 32512034 DOI: 10.1016/j.tox.2020.152506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/19/2020] [Accepted: 06/02/2020] [Indexed: 01/02/2023]
Abstract
Maternal nicotine exposure during pregnancy and lactation (NIC) is associated with dysfunction of white adipose tissue (WAT). We focused on the NIC-induced WAT angiogenesis and explored its sex and age differences. Pregnant rats were randomly assigned to NIC (1.0 mg/kg nicotine twice per day) or control groups. Distribution and density of blood vessels were observed. Angiogenesis-related genes were tested at 4, 12 and 26 weeks to estimate angiogenic activity. In vitro, nicotine concentration- and time-response experiments (0-50 μM) were conducted in 3T3-L1. Lipid accumulation and angiogenesis-related genes were tested. NIC increased the blood vessels in inguinal subcutaneous WAT (igSWAT) and gonadal WAT (gWAT) of 26-week-aged male and 4-week-aged female offspring. In males, nicotine showed higher angiogenic activity at 26 weeks than at 4 weeks in igSWAT and gWAT. In females, nicotine's angiogenic activity was higher at 4 weeks than 26 weeks in igSWAT and gWAT. In vitro, nicotine promoted adipocyte differentiation, and increased the expression of angiogenesis-related genes in concentration- and time dependent manners. In conclusion, NIC-induced enhancement of angiogenic activity in WAT presented sex and age differences: nicotine showed higher angiogenic activity in adulthood than in childhood of male offspring, but the converse results were observed in female offspring.
Collapse
Affiliation(s)
- Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China
| | - Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China
| | - Li Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China
| | - Li Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 185, DongHu Road, Wuhan, 430071, China.
| |
Collapse
|
19
|
Weekends-Off Lenvatinib for Unresectable Hepatocellular Carcinoma Improves Therapeutic Response and Tolerability toward Adverse Events. Cancers (Basel) 2020; 12:cancers12041010. [PMID: 32325921 PMCID: PMC7226076 DOI: 10.3390/cancers12041010] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/08/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Although lenvatinib has become the standard therapy for hepatocellular carcinoma (HCC), the high incidence rate of adverse events (AEs) is an issue. This study aimed to clarify the AEs of lenvatinib and the therapeutic impact of five days-on/two days-off administration (i.e., weekends-off strategy) for lenvatinib. Methods: We retrospectively assessed the therapeutic effects and AEs of 135 patients treated with lenvatinib, and the improvement of tolerability and therapeutic efficacy of 30 patients treated with the weekends-off strategy. We also evaluated lenvatinib-induced vascular changes in tumors and healthy organs using a mouse hepatoma model. Results: The incidence rates of any grade and grade ≥ 3 AEs were 82.1% and 49.6%. Fatigue was the most important AE since it resulted in dose reduction and discontinuation. Of the 30 patients who received weekends-off lenvatinib, 66.7% tolerated the AEs. Although 80.8% of the patients showed progression after dose reduction, the therapeutic response improved in 61.5% of the patients by weekends-off lenvatinib. Notably, weekends-off administration significantly prolonged the administration period and survival (p < 0.001 and p < 0.05). The mouse hepatoma model showed that weekends-off administration contributed to recovery of vascularity in the organs. Conclusion: Weekends-off administration of lenvatinib was useful to recover the therapeutic response and tolerability toward AEs.
Collapse
|
20
|
Zhang WX, Chen HJ, Fan J, Li GL, Sun A, Lan LY, Zhang L, Yan YE. The association between maternal nicotine exposure and adipose angiogenesis in female rat offspring: A mechanism of adipose tissue function changes. Toxicol Lett 2019; 318:12-21. [PMID: 31622651 DOI: 10.1016/j.toxlet.2019.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/04/2019] [Accepted: 10/12/2019] [Indexed: 02/08/2023]
Abstract
Maternal smoking during pregnancy and lactation is associated with increased fat mass in the offspring, but the mechanism by which this occurs is not fully understood. Our study focused on the relationships among maternal nicotine exposure, adipose angiogenesis and adipose tissue function in female offspring. Pregnant rats were randomly assigned to nicotine or control groups. Microvascular density, lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested in 4-, 12- and 26-week female offspring. In vitro, nicotine concentration- and time-response experiments were conducted in 3T3-L1. Lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested. The conditioned media of differentiated 3T3-L1 treated with nicotine were used to observe tube formation in human umbilical vein endothelial cells (HUVECs). Nicotine-exposed females presented higher adipose microvascular density. The gene expression of α7nAChR, Egr1 and FGF2 was significantly increased in gonadal white adipose tissue (gWAT) and inguinal subcutaneous WAT (igSWAT) of nicotine-exposed females at 4 weeks of age. The protein expression of α7nAChR, Egr1 and FGF2 was increased in gWAT and igSWAT of nicotine-exposed females at 4 weeks of age, and increased in gWAT at 26 weeks. In vitro, nicotine increased the expression of lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins in a concentration- and time-dependent manner. In the tube formation experiment, adipocytes affected by nicotine promoted HUVEC angiogenesis. Therefore, maternal nicotine exposure promoted the early angiogenesis of adipose tissue via the α7nAChR-Egr1-FGF2 signaling pathway, and this angiogenesis mechanism was associated with increased adipogenesis in adipose tissue of female offspring.
Collapse
Affiliation(s)
- Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Ao Sun
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Liu-Yi Lan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Li Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
21
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
22
|
Abstract
Development of novel and effective therapeutics for treating various cancers is probably the most congested and challenging enterprise of pharmaceutical companies. Diverse drugs targeting malignant and nonmalignant cells receive clinical approval each year from the FDA. Targeting cancer cells and nonmalignant cells unavoidably changes the tumor microenvironment, and cellular and molecular components relentlessly alter in response to drugs. Cancer cells often reprogram their metabolic pathways to adapt to environmental challenges and facilitate survival, proliferation, and metastasis. While cancer cells' dependence on glycolysis for energy production is well studied, the roles of adipocytes and lipid metabolic reprogramming in supporting cancer growth, metastasis, and drug responses are less understood. This Review focuses on emerging mechanisms involving adipocytes and lipid metabolism in altering the response to cancer treatment. In particular, we discuss mechanisms underlying cancer-associated adipocytes and lipid metabolic reprogramming in cancer drug resistance.
Collapse
|
23
|
Dipali SS, Ferreira CR, Zhou LT, Pritchard MT, Duncan FE. Histologic analysis and lipid profiling reveal reproductive age-associated changes in peri-ovarian adipose tissue. Reprod Biol Endocrinol 2019; 17:46. [PMID: 31189477 PMCID: PMC6563378 DOI: 10.1186/s12958-019-0487-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reproductive aging is a robust phenotype that occurs in all females and is characterized by a significant reduction in gamete quantity and quality, which can have negative consequences on both endocrine function and fertility. Age-associated differences in the oocyte, follicle, and ovary have been well-documented, but how the broader environment changes with age is less well understood. Fat is one of the largest organs in the body, and peri-gonadal adipose tissue surrounds the rodent ovary and comprises a local ovarian environment. The goal of this study was to characterize how peri-ovarian adipose tissue changes with advanced reproductive age. METHODS We isolated peri-gonadal adipose tissue from two cohorts of CB6F1 mice: reproductively young (6-12 weeks) and reproductively old (14-17 months). A comparative histological analysis was performed to evaluate adipocyte architecture. We then extracted lipids from the tissue and performed multiple reaction monitoring (MRM)-profiling, a mass spectrometry-based method of metabolite profiling, to compare the lipid profiles of peri-gonadal adipose tissue in these age cohorts. RESULTS We found that advanced reproductive age was associated with adipocyte hypertrophy and a corresponding decrease in the number of adipocytes per area. Of the 10 lipid classes examined, triacylglycerols (TAGs) had significantly different profiles between young and old cohorts, despite quantitative analysis revealing a decrease in the total amount of TAGs per weight of peri-gonadal adipose tissue with age. CONCLUSIONS These findings pinpoint age-associated physiological changes in peri-gonadal adipose tissue with respect to adipocyte morphology and lipid profiles and lay the foundation for future studies to examine how these alterations may influence both adipocyte and ovarian function.
Collapse
Affiliation(s)
- Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Christina R Ferreira
- Center for Analytical Instrumentation Development, Department of Chemistry, Purdue University, West Lafayette, IN, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA.
| |
Collapse
|
24
|
Chen Y, Zhao M, Zheng T, Adlat S, Jin H, Wang C, Li D, Zaw Myint MZ, Yao Y, Xu L, San M, Wen H, Zhang Y, Lu X, Yang L, Zhang L, Feng X, Zheng Y. Repression of adipose vascular endothelial growth factor reduces obesity through adipose browning. Am J Physiol Endocrinol Metab 2019; 316:E145-E155. [PMID: 30398903 DOI: 10.1152/ajpendo.00196.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity is the result of excessive energy accumulation and is associated with many diseases. We previously reported that universal repression of vascular endothelial growth factor (VEGF) leads to brown-like adipocyte development in white adipose tissues, and that these mice are resistant to obesity (Lu X et al. Endocrinology 153: 3123-3132, 2012). Using an adipose-specific VEGF repression mouse model (aP2-rtTR-krabtg/+/VEGFtetO/tetO), we show that adipose-specific VEGF repression can repeat the previous phenotypes, including adipose browning, increased energy consumption, and reduction in body weight. Expression of brown adipose-associated genes is increased, and white adipose-associated genes are downregulated under VEGF repression. Our study demonstrates that adipose-specific VEGF repression can lead to antiobesity activity through adipose browning and has potential clinical value.
Collapse
Affiliation(s)
- Yang Chen
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Mingyue Zhao
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Tingting Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Salah Adlat
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Honghong Jin
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Chenhao Wang
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Dan Li
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - May Zun Zaw Myint
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Yapeng Yao
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Liu Xu
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Mingjun San
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Huaizhen Wen
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Yuntao Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Xiaodan Lu
- Transgenic Research Center, Northeast Normal University, Changchun, China
| | - Ling Yang
- Shanxi Medical University , Taiyuan , China
| | - Luqing Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University , Changchun , China
| | - Xuechao Feng
- Transgenic Research Center, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University , Changchun , China
| | - Yaowu Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University , Changchun , China
| |
Collapse
|
25
|
Graupera M, Claret M. Endothelial Cells: New Players in Obesity and Related Metabolic Disorders. Trends Endocrinol Metab 2018; 29:781-794. [PMID: 30266200 DOI: 10.1016/j.tem.2018.09.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Abstract
Metabolic disorders such as obesity are accompanied by endothelial cell (EC) dysfunction and decreased vascular density. The current paradigm posits that metabolic alterations associated with obesity secondarily lead to EC dysfunction. However, in view of recent evidence reporting that EC dysfunction per se is able to cause metabolic dysregulation, this paradigm should be revisited and further elaborated. In this article we summarize current views and discuss evidence in favor of a causal role for ECs in systemic metabolic dysregulation. We also integrate and contextualize current research in a pathophysiological framework and discuss potential therapeutic strategies targeting angiogenesis to help to counteract obesity.
Collapse
Affiliation(s)
- Mariona Graupera
- Vascular Signaling Laboratory, ProCURE and Oncobell Programs, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 l'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain.
| |
Collapse
|
26
|
Iwamoto H, Abe M, Yang Y, Cui D, Seki T, Nakamura M, Hosaka K, Lim S, Wu J, He X, Sun X, Lu Y, Zhou Q, Shi W, Torimura T, Nie G, Li Q, Cao Y. Cancer Lipid Metabolism Confers Antiangiogenic Drug Resistance. Cell Metab 2018; 28:104-117.e5. [PMID: 29861385 DOI: 10.1016/j.cmet.2018.05.005] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 11/02/2017] [Accepted: 05/03/2018] [Indexed: 01/01/2023]
Abstract
Intrinsic and evasive antiangiogenic drug (AAD) resistance is frequently developed in cancer patients, and molecular mechanisms underlying AAD resistance remain largely unknown. Here we describe AAD-triggered, lipid-dependent metabolic reprogramming as an alternative mechanism of AAD resistance. Unexpectedly, tumor angiogenesis in adipose and non-adipose environments is equally sensitive to AAD treatment. AAD-treated tumors in adipose environment show accelerated growth rates in the presence of a minimal number of microvessels. Mechanistically, AAD-induced tumor hypoxia initiates the fatty acid oxidation metabolic reprogramming and increases uptake of free fatty acid (FFA) that stimulates cancer cell proliferation. Inhibition of carnitine palmitoyl transferase 1A (CPT1) significantly compromises the FFA-induced cell proliferation. Genetic and pharmacological loss of CPT1 function sensitizes AAD therapeutic efficacy and enhances its anti-tumor effects. Together, we propose an effective cancer therapy concept by combining drugs that target angiogenesis and lipid metabolism.
Collapse
Affiliation(s)
- Hideki Iwamoto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden; Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Mitsuhiko Abe
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden; Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Dongmei Cui
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Masaki Nakamura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Sharon Lim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Jieyu Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Xingkang He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Xiaoting Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden; Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yongtian Lu
- Key Laboratory of International Collaborations, Second People's Hospital of Shenzhen, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Yanerdao Road, Qingdao 266071, China
| | - Weiyun Shi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Yanerdao Road, Qingdao 266071, China
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Guohui Nie
- Key Laboratory of International Collaborations, Second People's Hospital of Shenzhen, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 171 77, Sweden.
| |
Collapse
|
27
|
Cao Y, Wang H, Wang Q, Han X, Zeng W. Three-dimensional volume fluorescence-imaging of vascular plasticity in adipose tissues. Mol Metab 2018; 14:71-81. [PMID: 29914852 PMCID: PMC6034070 DOI: 10.1016/j.molmet.2018.06.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/16/2018] [Accepted: 06/02/2018] [Indexed: 01/16/2023] Open
Abstract
Objective The vascular system is central to sustaining tissue survival and homeostasis. Blood vessels are densely present in adipose tissues and exert essential roles in their metabolism. However, conventional immunohistochemistry methods have intrinsic limitations in examining the 3D vascular network in adipose tissues as well as other organs in general. Methods We established a 3D volume fluorescence-imaging technique to visualize the vasculatures in mouse adipose tissues by combining the optimized steps of whole-mount immunolabeling, tissue optical clearing, and lightsheet volume fluorescence-imaging. To demonstrate the strength of this novel imaging procedure, we comprehensively assessed the intra-adipose vasculatures under obese conditions or in response to a cold challenge. Results We show the entirety of the vascular network in mouse adipose tissues on the whole-tissue level at a single-capillary resolution for the first time in the field. We accurately quantify the pathological changes of vasculatures in adipose tissues in wild-type or obese mice (ob/ob, db/db, or diet-induced obesity). In addition, we identify significant and reversible changes of the intra-adipose vasculatures in the mice subjected to cold challenge (i.e., 4°). Furthermore, we demonstrate that the cold-induced vascular plasticity depends on the sympathetic-derived catecholamine signal and is involved in the beiging process of white adipose tissues. Conclusions We report a 3D volume fluorescence-imaging procedure that is compatible with many areas of vascular research and is poised to serve the field in future investigations of the vascular system in adipose tissues or other research scenarios. 3D vascular network in adipose tissues is visualized at single-capillary resolution. Pathological remodeling of vasculatures is characterized under the obese conditions. Vascular plasticity during cold challenge is involved in the beiging process of WAT. Sympathetic-derived catecholamine signal regulates the vascular plasticity in WAT.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Tsinghua University, Beijing, 100084, China; Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Huanhuan Wang
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Qi Wang
- Center for Life Sciences, Tsinghua University, Beijing, 100084, China; Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Xiangli Han
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Wenwen Zeng
- Center for Life Sciences, Tsinghua University, Beijing, 100084, China; Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China.
| |
Collapse
|
28
|
Li J, Yang Q, Bai Z, Zhou W, Semenza GL, Ge RL. Chronic cold exposure results in subcutaneous adipose tissue browning and altered global metabolism in Qinghai-Tibetan plateau pika (Ochotona curzoniae). Biochem Biophys Res Commun 2018; 500:117-123. [PMID: 29626477 DOI: 10.1016/j.bbrc.2018.03.147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 12/15/2022]
Abstract
The plateau pika (Ochotona curzoniae), one of the indigenous animals of the Qinghai-Tibet Plateau, is adapted to life in a cold and hypoxic environment. We conducted a series of genomic, proteomic and morphological studies to investigate whether changes in energy metabolism contribute to adaptation of the plateau pika to cold stress by analyzing summer and winter cohorts. The winter group showed strong morphological and histological features of brown adipose tissue (BAT) in subcutaneous white adipose tissue (sWAT). To obtain molecular evidence of browning of sWAT, we performed reverse transcription and quantitative real-time PCR, which revealed that BAT-specific genes, including uncoupling protein 1 (UCP-1) and PPAR-γ coactivator 1α (PGC-1α), were highly expressed in sWAT from the winter group. Compared with the summer group, Western blot analysis also confirmed that UCP-1, PGC-1α and Cox4 protein levels were significantly increased in sWAT from the winter group. Increased BAT mass in the inter-scapular region of the winter group was also observed. These results suggest that the plateau pika adapts to cold by browning sWAT and increasing BAT in order to increase thermogenesis. These changes are distinct from the previously reported adaptation of highland deer mice. Understanding the regulatory mechanisms underlying this adaptation may lead to novel therapeutic strategies for treating obesity and metabolic disorders.
Collapse
Affiliation(s)
- Jia Li
- Research Center for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China
| | - Quanyu Yang
- Research Center for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China
| | - Zhenzhong Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China.
| | - Wenhua Zhou
- Research Center for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China
| | - Gregg L Semenza
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University Medical College, 810001 Qinghai, Xining, PR China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Qinghai University Medical College, 810001 Qinghai, Xining, PR China.
| |
Collapse
|
29
|
Seki T, Hosaka K, Fischer C, Lim S, Andersson P, Abe M, Iwamoto H, Gao Y, Wang X, Fong GH, Cao Y. Ablation of endothelial VEGFR1 improves metabolic dysfunction by inducing adipose tissue browning. J Exp Med 2018; 215:611-626. [PMID: 29305395 PMCID: PMC5789413 DOI: 10.1084/jem.20171012] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/19/2017] [Accepted: 12/05/2017] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)-fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.
Collapse
Affiliation(s)
- Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Carina Fischer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Sharon Lim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Patrik Andersson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mitsuhiko Abe
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Hideki Iwamoto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Yanyan Gao
- Central Research Laboratory, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinsheng Wang
- Central Research Laboratory, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guo-Hua Fong
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden .,Central Research Laboratory, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
30
|
Oubaha M, Miloudi K, Dejda A, Guber V, Mawambo G, Germain MA, Bourdel G, Popovic N, Rezende FA, Kaufman RJ, Mallette FA, Sapieha P. Senescence-associated secretory phenotype contributes to pathological angiogenesis in retinopathy. Sci Transl Med 2017; 8:362ra144. [PMID: 27797960 DOI: 10.1126/scitranslmed.aaf9440] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/17/2016] [Indexed: 12/13/2022]
Abstract
Pathological angiogenesis is the hallmark of diseases such as cancer and retinopathies. Although tissue hypoxia and inflammation are recognized as central drivers of vessel growth, relatively little is known about the process that bridges the two. In a mouse model of ischemic retinopathy, we found that hypoxic regions of the retina showed only modest rates of apoptosis despite severely compromised metabolic supply. Using transcriptomic analysis and inducible loss-of-function genetics, we demonstrated that ischemic retinal cells instead engage the endoplasmic reticulum stress inositol-requiring enzyme 1α (IRE1α) pathway that, through its endoribonuclease activity, induces a state of senescence in which cells adopt a senescence-associated secretory phenotype (SASP). We also detected SASP-associated cytokines (plasminogen activator inhibitor 1, interleukin-6, interleukin-8, and vascular endothelial growth factor) in the vitreous humor of patients suffering from proliferative diabetic retinopathy. Therapeutic inhibition of the SASP through intravitreal delivery of metformin or interference with effectors of senescence (semaphorin 3A or IRE1α) in mice reduced destructive retinal neovascularization in vivo. We conclude that the SASP contributes to pathological vessel growth, with ischemic retinal cells becoming prematurely senescent and secreting inflammatory cytokines that drive paracrine senescence, exacerbate destructive angiogenesis, and hinder reparative vascular regeneration. Reversal of this process may be therapeutically beneficial.
Collapse
Affiliation(s)
- Malika Oubaha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada
| | - Khalil Miloudi
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Vera Guber
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Gaëlle Mawambo
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Marie-Anne Germain
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada.,Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Guillaume Bourdel
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Natalija Popovic
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Flavio A Rezende
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Frédérick A Mallette
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada. .,Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| |
Collapse
|
31
|
Ma L, Ni M, Hao P, Lu H, Yang X, Xu X, Zhang C, Huang S, Zhao Y, Liu X, Zhang Y. Tongxinluo mitigates atherogenesis by regulating angiogenic factors and inhibiting vasa vasorum neovascularization in apolipoprotein E-deficient mice. Oncotarget 2017; 7:16194-204. [PMID: 26908443 PMCID: PMC4941307 DOI: 10.18632/oncotarget.7477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/08/2016] [Indexed: 02/05/2023] Open
Abstract
Vasa vasorum (VV) neovascularization contributes to atherogenesis and its expansion and distribution is correlated with intraplaque expression of angiogenic factors. The present study investigated the roles of Tongxinluo (TXL), a traditional Chinese medication, on VV proliferation and atherogenesis. In vitro, TXL pre-treatment reversed the tumor necrosis factor-a (TNF-a) induced expression of vascular endothelial growth factor A (VEGF-A) and angiopoietin-1 (ANGPT-1) but not ANGPT-2, leading to increased ratio of ANGPT-1 to ANGPT-2. Consistently, TXL treatment (at a dosage of 0.38, 0.75, 1.5 g/kg/d, respectively) decreased the expression of VEGF-A while increased that of ANGPT-1 in early atherosclerotic lesions of apolipoprotein E deficient (apoE−/−) mice. On aortic ring assay, microvessels sprouting from aortas were significantly inhibited in TXL-treated mice. Moreover, VV neovascularization in plaques was markedly reduced with TXL treatment. Histological and morphological analysis demonstrated that TXL treatment reduced plaque burden, plaque size and changed the plaque composition. These data suggest that TXL inhibits early atherogenesis through regulating angiogenic factor expression and inhibiting VV proliferation in atherosclerotic plaque. Our study shed new light on the anti-atherosclerotic effect of TXL.
Collapse
Affiliation(s)
- Lianyue Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xiaoyan Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xingli Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Shanying Huang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Yuxia Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| |
Collapse
|
32
|
Luque RM, López-Sánchez LM, Villa-Osaba A, Luque IM, Santos-Romero AL, Yubero-Serrano EM, Cara-García M, Álvarez-Benito M, López-Mirand a J, Gahete MD, Castaño JP. Breast cancer is associated to impaired glucose/insulin homeostasis in premenopausal obese/overweight patients. Oncotarget 2017; 8:81462-81474. [PMID: 29113405 PMCID: PMC5655300 DOI: 10.18632/oncotarget.20399] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/25/2017] [Indexed: 01/04/2023] Open
Abstract
The association between breast cancer (BCa) presence and altered glucose/insulin metabolism is controversial likely due to an inaccurate insulin resistance (IR) assessment and inappropriate stratification of patients by body-mass index (BMI) and menopausal state. 148 women with suspect of sporadic BCa were stratified by BMI and menopause. Fasting levels of glucose, insulin, glycohemoglobin and selected IR-related and tumor-derived markers were measured. Glucose/insulin levels during OGTT were used to calculate insulin resistance/sensitivity indexes. Analysis of 77 BCa-bearing patients and 71 controls showed an association between BCa and IR as demonstrated by impaired glucose/insulin homeostasis (increased fasting- and OGTT-induced glucose levels) and deteriorated IR indexes, which was especially patent in premenopausal women. The association between BCa presence and IR was markedly influenced by BMI, being obese BCa patients significantly more insulin resistant than controls. BCa presence was associated to elevated levels of IR (glucose, triglycerides) and tumor-derived (VEGF) markers, especially in overweight/obese patients. BCa presence is associated to IR in overweight/obese premenopausal but not in premenopausal normal weight or postmenopausal women. Our data support a bidirectional relationship between dysregulated/imbalanced glucose/insulin metabolism and BCa, as tumor- and IR-markers are correlated with the impairment of glucose/insulin metabolism in overweight/obese premenopausal BCa patients.
Collapse
Affiliation(s)
- Raúl M. Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
- CIBERobn, Córdoba, Spain
- ceiA3, Córdoba, Spain
| | - Laura M. López-Sánchez
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
- CIBERobn, Córdoba, Spain
- ceiA3, Córdoba, Spain
| | - Alicia Villa-Osaba
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
- CIBERobn, Córdoba, Spain
- ceiA3, Córdoba, Spain
| | - Isabel M. Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Ana L. Santos-Romero
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBERobn, Córdoba, Spain
- Mammary Gland Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Elena M. Yubero-Serrano
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBERobn, Córdoba, Spain
- Lipids and Atherosclerosis Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - María Cara-García
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBERobn, Córdoba, Spain
- Mammary Gland Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Marina Álvarez-Benito
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBERobn, Córdoba, Spain
- Mammary Gland Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - José López-Mirand a
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBERobn, Córdoba, Spain
- Lipids and Atherosclerosis Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Manuel D. Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
- CIBERobn, Córdoba, Spain
- ceiA3, Córdoba, Spain
| | - Justo P. Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofia, Córdoba, Spain
- CIBERobn, Córdoba, Spain
- ceiA3, Córdoba, Spain
| |
Collapse
|
33
|
Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev 2017; 274:307-329. [PMID: 27782324 DOI: 10.1111/imr.12479] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelium is strategically located at the interface between blood and interstitial tissues, placing thus endothelial cell as a key player in vascular homeostasis. Endothelial cells are in a dynamic equilibrium with their environment and constitute concomitantly a source, a barrier, and a target of defensive mediators. This review will discuss the recent advances in our understanding of the complex crosstalk between the endothelium, the complement system and the hemostasis in health and in disease. The first part will provide a general introduction on endothelial cells heterogeneity and on the physiologic role of the complement and hemostatic systems. The second part will analyze the interplay between complement, hemostasis and endothelial cells in physiological conditions and their alterations in diseases. Particular focus will be made on the prototypes of thrombotic microangiopathic disorders, resulting from complement or hemostasis dysregulation-mediated endothelial damage: atypical hemolytic uremic syndrome and thrombotic thrombocytopenic purpura. Novel aspects of the pathophysiology of the thrombotic microangiopathies will be discussed.
Collapse
Affiliation(s)
- Lubka T Roumenina
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Marie Frimat
- INSERM UMR 995, Lille, France.,Nephrology Department, CHU Lille, Lille, France
| | - Veronique Fremeaux-Bacchi
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
34
|
Maintenance of antiangiogenic and antitumor effects by orally active low-dose capecitabine for long-term cancer therapy. Proc Natl Acad Sci U S A 2017; 114:E5226-E5235. [PMID: 28607065 DOI: 10.1073/pnas.1705066114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Long-term uninterrupted therapy is essential for maximizing clinical benefits of antiangiogenic drugs (AADs) in cancer patients. Unfortunately, nearly all clinically available AADs are delivered to cancer patients using disrupted regimens. We aim to develop lifetime, nontoxic, effective, orally active, and low-cost antiangiogenic and antitumor drugs for treatment of cancer patients. Here we report our findings of long-term maintenance therapy with orally active, nontoxic, low cost antiangiogenic chemotherapeutics for effective cancer treatment. In a sequential treatment regimen, robust antiangiogenic effects in tumors were achieved with an anti-VEGF drug, followed by a low-dose chemotherapy. The nontoxic, low dose of the orally active prodrug capecitabine was able to sustain the anti-VEGF-induced vessel regression for long periods. In another experimental setting, maintenance of low-dose capecitabine produced greater antiangiogenic and antitumor effects after AAD plus chemotherapy. No obvious adverse effects were developed after more than 2-mo of consecutive treatment with a low dose of capecitabine. Together, our findings provide a rationalized concept of effective cancer therapy by long-term maintenance of AAD-triggered antiangiogenic effects with orally active, nontoxic, low-cost, clinically available chemotherapeutics.
Collapse
|
35
|
Park J, Kim M, Sun K, An YA, Gu X, Scherer PE. VEGF-A-Expressing Adipose Tissue Shows Rapid Beiging and Enhanced Survival After Transplantation and Confers IL-4-Independent Metabolic Improvements. Diabetes 2017; 66:1479-1490. [PMID: 28254844 PMCID: PMC5440018 DOI: 10.2337/db16-1081] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 02/23/2017] [Indexed: 01/01/2023]
Abstract
Adipocyte-derived vascular endothelial growth factor-A (VEGF-A) plays a crucial role in angiogenesis and contributes to adipocyte function and systemic metabolism, such as insulin resistance, chronic inflammation, and beiging of subcutaneous adipose tissue. Using a doxycycline-inducible adipocyte-specific VEGF-A-overexpressing mouse model, we investigated the dynamics of local VEGF-A effects on tissue beiging of adipose tissue transplants. VEGF-A overexpression in adipocytes triggers angiogenesis. We also observed a rapid appearance of beige fat cells in subcutaneous white adipose tissue as early as 2 days postinduction of VEGF-A. In contrast to conventional cold-induced beiging, VEGF-A-induced beiging is independent of interleukin-4. We subjected metabolically healthy VEGF-A-overexpressing adipose tissue to autologous transplantation. Transfer of subcutaneous adipose tissues taken from VEGF-A-overexpressing mice into diet-induced obese mice resulted in systemic metabolic benefits, associated with improved survival of adipocytes and a concomitant reduced inflammatory response. These effects of VEGF-A are tissue autonomous, inducing white adipose tissue beiging and angiogenesis within the transplanted tissue. Our findings indicate that manipulation of adipocyte functions with a bona fide angiogenic factor, such as VEGF-A, significantly improves the survival and volume retention of fat grafts and can convey metabolically favorable properties on the recipient on the basis of beiging.
Collapse
Affiliation(s)
- Jiyoung Park
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Min Kim
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Inje University, and Cardiovascular and Metabolic Disease Center, Inje University, Busanjin-gu, Busan, South Korea
| | - Kai Sun
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX
| | - Yu Aaron An
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Xue Gu
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX
| | - Philipp E Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
36
|
Frueh FS, Später T, Scheuer C, Menger MD, Laschke MW. Isolation of Murine Adipose Tissue-derived Microvascular Fragments as Vascularization Units for Tissue Engineering. J Vis Exp 2017. [PMID: 28518106 DOI: 10.3791/55721] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A functional microvascular network is of pivotal importance for the survival and integration of engineered tissue constructs. For this purpose, several angiogenic and prevascularization strategies have been established. However, most cell-based approaches include time-consuming in vitro steps for the formation of a microvascular network. Hence, they are not suitable for intraoperative one-step procedures. Adipose tissue-derived microvascular fragments (ad-MVF) represent promising vascularization units. They can be easily isolated from fat tissue and exhibit a functional microvessel morphology. Moreover, they rapidly reassemble into new microvascular networks after in vivo implantation. In addition, ad-MVF have been shown to induce lymphangiogenesis. Finally, they are a rich source of mesenchymal stem cells, which may further contribute to their high vascularization potential. In previous studies we have demonstrated the remarkable vascularization capacity of ad-MVF in engineered bone and skin substitutes. In the present study, we report on a standardized protocol for the enzymatic isolation of ad-MVF from murine fat tissue.
Collapse
Affiliation(s)
- Florian S Frueh
- Institute for Clinical and Experimental Surgery, Saarland University; Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, University of Zurich;
| | - Thomas Später
- Institute for Clinical and Experimental Surgery, Saarland University
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University
| | | |
Collapse
|
37
|
Wang Y, Lin M, Gao X, Pedram P, Du J, Vikram C, Gulliver W, Zhang H, Sun G. High dietary selenium intake is associated with less insulin resistance in the Newfoundland population. PLoS One 2017; 12:e0174149. [PMID: 28380029 PMCID: PMC5381811 DOI: 10.1371/journal.pone.0174149] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/03/2017] [Indexed: 02/07/2023] Open
Abstract
As an essential nutrient, Selenium (Se) is involved in many metabolic activities including mimicking insulin function. Data on Se in various biological samples and insulin resistance are contradictory, moreover there is no large study available regarding the relationship of dietary Se intake with insulin resistance in the general population. To investigate the association between dietary Se intake and variation of insulin resistance in a large population based study, a total of 2420 subjects without diabetes from the CODING (Complex Diseases in the Newfoundland Population: Environment and Genetics) study were assessed. Dietary Se intake was evaluated from the Willett Food Frequency questionnaire. Fasting blood samples were used for the measurement of glucose and insulin. Insulin resistance was determined with the homeostasis model assessment (HOMA-IR). Body composition was measured using dual energy X-ray absorptiometry. Analysis of covariance showed that high HOMA-IR groups in both males and females had the lowest dietary Se intake (μg/kg/day) (p < 0.01), being 18% and 11% lower than low HOMA-IR groups respectively. Insulin resistance decreased with the increase of dietary Se intake in females but not in males after controlling for age, total calorie intake, physical activity level, serum calcium, serum magnesium, and body fat percentage (p < 0.01). Partial correlation analysis showed that dietary Se intake was negatively correlated with HOMA-IR after adjusting for the Se confounding factors in subjects whose dietary Se intake was below 1.6 μg/kg/day (r = -0.121 for males and -0.153 for females, p < 0.05). However, the negative correlation was no longer significant when dietary Se intake was above 1.6 μg/kg/day. Our findings suggest that higher dietary Se intake is beneficially correlated with lower insulin resistance when total dietary Se intake was below 1.6 μg/kg/day. Above this cutoff, this beneficial effect disappears.
Collapse
Affiliation(s)
- Yongbo Wang
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Meiju Lin
- Department of Biliary Minimally Invasive Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiang Gao
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Pardis Pedram
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Jianling Du
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chandurkar Vikram
- Division of Endocrinology, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Wayne Gulliver
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Hongwei Zhang
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Guang Sun
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| |
Collapse
|
38
|
Knockout of Vasohibin-1 Gene in Mice Results in Healthy Longevity with Reduced Expression of Insulin Receptor, Insulin Receptor Substrate 1, and Insulin Receptor Substrate 2 in Their White Adipose Tissue. J Aging Res 2017; 2017:9851380. [PMID: 28367331 PMCID: PMC5358453 DOI: 10.1155/2017/9851380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/30/2017] [Accepted: 02/16/2017] [Indexed: 12/27/2022] Open
Abstract
Vasohibin-1 (Vash1), originally isolated as an endothelium-derived angiogenesis inhibitor, has a characteristic of promoting stress tolerance in endothelial cells (ECs). We therefore speculated that the lack of the vash1 gene would result in a short lifespan. However, to our surprise, vash1−/− mice lived significantly longer with a milder senescence phenotype than wild-type (WT) mice. We sought the cause of this healthy longevity and found that vash1−/− mice exhibited mild insulin resistance along with reduced expression of the insulin receptor (insr), insulin receptor substrate 1 (irs-1), and insulin receptor substrate 2 (irs-2) in their white adipose tissue (WAT) but not in their liver or skeletal muscle. The expression of vash1 dominated in the WAT among those 3 organs. Importantly, vash1−/− mice did not develop diabetes even when fed a high-fat diet. These results indicate that the expression of vash1 was required for the normal insulin sensitivity of the WAT and that the target molecules for this activity were insr, irs1, and irs2. The lack of vash1 caused mild insulin resistance without the outbreak of overt diabetes and might contribute to healthy longevity.
Collapse
|
39
|
Sathyapalan T, Javed Z, Kilpatrick ES, Coady AM, Atkin SL. Endocannabinoid receptor blockade increases vascular endothelial growth factor and inflammatory markers in obese women with polycystic ovary syndrome. Clin Endocrinol (Oxf) 2017; 86:384-387. [PMID: 27651218 DOI: 10.1111/cen.13239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/17/2016] [Accepted: 09/17/2016] [Indexed: 12/25/2022]
Abstract
CONTEXT Animal studies suggest that cannabinoid receptor-1 (CB-1) blockade reduces inflammation and neovascularization by decreasing vascular endothelial growth factor (VEGF) levels associated with a reduction in inflammatory markers, thereby potentially reducing cardiovascular risk. OBJECTIVE To determine the impact of CB1 antagonism by rimonabant on VEGF and inflammatory markers in obese PCOS women. DESIGN Randomized, open-labelled parallel study. SETTING Endocrinology outpatient clinic in a referral centre. SUBJECTS Twenty patients with PCOS (PCOS) and biochemical hyperandrogenaemia with a body mass index of ≥30 kg/m2 were recruited. Patients were randomized to 1·5 g daily of metformin or 20 mg daily of rimonabant. MAIN OUTCOME MEASURES Post hoc review to detect VEGF and pro-inflammatory cytokines TNF-α, IL-1β, IL-1ra, IL-2, IL6, IL-8, IL-10 and MCP-1 before and after 12 weeks of treatment. RESULTS After 12 weeks of rimonabant treatment, there was a significant increase in VEGF (99·2 ± 17·6 vs 116·2 ± 15·8 pg/ml, P < 0·01) and IL-8 (7·4 ± 11·0 vs 18·1 ± 13·2 pg/ml, P < 0·05) but not after metformin (VEGF P = 0·7; IL-8 P = 0·9). There was no significant difference in the pro-inflammatory cytokines TNF-α, IL-1β, IL-1ra, IL-2, IL6, IL-8, IL-10 and MCP-1 following either treatment. CONCLUSION This study suggests that rimonabant CB-I blockade paradoxically raised VEGF and the cytokine IL-8 in obese women with PCOS that may have offset the potential benefit associated with weight loss.
Collapse
Affiliation(s)
- Thozhukat Sathyapalan
- Department of Academic Endocrinology, Diabetes and Metabolism, University of Hull, Hull, UK
| | - Zeeshan Javed
- Department of Academic Endocrinology, Diabetes and Metabolism, University of Hull, Hull, UK
| | | | - Anne-Marie Coady
- Department of Obstetric Ultrasound, Hull & East Yorkshire Women's & Children's Hospital, Hull, UK
| | | |
Collapse
|
40
|
Yang X, Sui W, Zhang M, Dong M, Lim S, Seki T, Guo Z, Fischer C, Lu H, Zhang C, Yang J, Zhang M, Wang Y, Cao C, Gao Y, Zhao X, Sun M, Sun Y, Zhuang R, Samani NJ, Zhang Y, Cao Y. Switching harmful visceral fat to beneficial energy combustion improves metabolic dysfunctions. JCI Insight 2017; 2:e89044. [PMID: 28239649 PMCID: PMC5313060 DOI: 10.1172/jci.insight.89044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/05/2017] [Indexed: 01/16/2023] Open
Abstract
Visceral fat is considered the genuine and harmful white adipose tissue (WAT) that is associated to development of metabolic disorders, cardiovascular disease, and cancer. Here, we present a new concept to turn the harmful visceral fat into a beneficial energy consumption depot, which is beneficial for improvement of metabolic dysfunctions in obese mice. We show that low temperature-dependent browning of visceral fat caused decreased adipose weight, total body weight, and body mass index, despite increased food intake. In high-fat diet-fed mice, low temperature exposure improved browning of visceral fat, global metabolism via nonshivering thermogenesis, insulin sensitivity, and hepatic steatosis. Genome-wide expression profiling showed upregulation of WAT browning-related genes including Cidea and Dio2. Conversely, Prdm16 was unchanged in healthy mice or was downregulated in obese mice. Surgical removal of visceral fat and genetic knockdown of UCP1 in epididymal fat largely ablated low temperature-increased global thermogenesis and resulted in the death of most mice. Thus, browning of visceral fat may be a compensatory heating mechanism that could provide a novel therapeutic strategy for treating visceral fat-associated obesity and diabetes.
Collapse
Affiliation(s)
- Xiaoyan Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wenhai Sui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sharon Lim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ziheng Guo
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Carina Fischer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Yangang Wang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Caixia Cao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanyan Gao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xingguo Zhao
- Department of Otolaryngology, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Meili Sun
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
| | - Rujie Zhuang
- The TCM Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | - Yihai Cao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| |
Collapse
|
41
|
Takeda E, Suzuki Y, Sato Y. Age-associated downregulation of vasohibin-1 in vascular endothelial cells. Aging Cell 2016; 15:885-92. [PMID: 27325558 PMCID: PMC5013028 DOI: 10.1111/acel.12497] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 12/21/2022] Open
Abstract
Vasohibin-1 (VASH1) is an angiogenesis-inhibiting factor synthesized by endothelial cells (ECs) and it also functions to increase stress tolerance of ECs, which function is critical for the maintenance of vascular integrity. Here, we examined whether the expression of VASH1 would be affected by aging. We passaged human umbilical vein endothelial cells (HUVECs) and observed that VASH1 was downregulated in old HUVECs. This decrease in VASH1 expression with aging was confirmed in mice. To explore the mechanism of this downregulation, we compared the expression of microRNAs between old and young HUVECs by performing microarray analysis. Among the top 20 microRNAs that were expressed at a higher level in old HUVECs, the third highest microRNA, namely miR-22-3p, had its binding site on the 3' UTR of VASH1 mRNA. Experiments with microRNA mimic and anti-miR revealed that miR-22-3p was involved at least in part in the downregulation of VASH1 in ECs during replicative senescence. We then clarified the significance of this defective expression of VASH1 in the vasculature. When a cuff was placed around the femoral arteries of wild-type mice and VASH1-null mice, neointimal formation was augmented in the VASH1-null mice accompanied by an increase in adventitial angiogenesis, macrophage accumulation in the adventitia, and medial/neointimal proliferating cells. These results indicate that in replicative senescence, the downregulation of VASH1 expression in ECs was caused, at least in part, by the alteration of microRNA expression. Such downregulation of VASH1 might be involved in the acceleration of age-associated vascular diseases.
Collapse
Affiliation(s)
- Eichi Takeda
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| | - Yasuhiro Suzuki
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| | - Yasufumi Sato
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| |
Collapse
|
42
|
Discontinuation of anti-VEGF cancer therapy promotes metastasis through a liver revascularization mechanism. Nat Commun 2016; 7:12680. [PMID: 27580750 PMCID: PMC5025794 DOI: 10.1038/ncomms12680] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/21/2016] [Indexed: 12/21/2022] Open
Abstract
The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the ‘off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers. Anti-VEGF therapy often produces limited beneficial effects in cancer patients. Here, the authors show that discontinuation of anti-VEGF cancer therapy in xenografts-bearing mice increases cancer cells extravasation and intravasation in liver through the host-derived VEGF.
Collapse
|
43
|
Seki T, Hosaka K, Lim S, Fischer C, Honek J, Yang Y, Andersson P, Nakamura M, Näslund E, Ylä-Herttuala S, Sun M, Iwamoto H, Li X, Liu Y, Samani NJ, Cao Y. Endothelial PDGF-CC regulates angiogenesis-dependent thermogenesis in beige fat. Nat Commun 2016; 7:12152. [PMID: 27492130 PMCID: PMC4980448 DOI: 10.1038/ncomms12152] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/06/2016] [Indexed: 12/18/2022] Open
Abstract
Cold- and β3-adrenoceptor agonist-induced sympathetic activation leads to angiogenesis and UCP1-dependent thermogenesis in mouse brown and white adipose tissues. Here we show that endothelial production of PDGF-CC during white adipose tissue (WAT) angiogenesis regulates WAT browning. We find that genetic deletion of endothelial VEGFR2, knockout of the Pdgf-c gene or pharmacological blockade of PDGFR-α impair the WAT-beige transition. We further show that PDGF-CC stimulation upregulates UCP1 expression and acquisition of a beige phenotype in differentiated mouse WAT-PDGFR-α+ progenitor cells, as well as in human WAT-PDGFR-α+ adipocytes, supporting the physiological relevance of our findings. Our data reveal a paracrine mechanism by which angiogenic endothelial cells modulate adipocyte metabolism, which may provide new targets for the treatment of obesity and related metabolic diseases. Cold-induced activation of thermogenesis in white adipose tissue (WAT), or ‘beiging', is associated with WAT angiogenesis. Here the authors show that PDGF-CC is secreted from endothelial cells in the context of WAT angiogenesis and its paracrine action on adipocytes contributes to cold-induced beiging.
Collapse
Affiliation(s)
- Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Sharon Lim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Carina Fischer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Jennifer Honek
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Yunlong Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Patrik Andersson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Masaki Nakamura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Erik Näslund
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm 182 88, Sweden
| | - Seppo Ylä-Herttuala
- Department of Molecular Medicine, A.I. Virtanen Institute, Molecular Sciences University of Eastern Finland, Kuopio 70211, Finland
| | - Meili Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Hideki Iwamoto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden.,Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
44
|
Falcon BL, Chintharlapalli S, Uhlik MT, Pytowski B. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents. Pharmacol Ther 2016; 164:204-25. [PMID: 27288725 DOI: 10.1016/j.pharmthera.2016.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interaction of numerous signaling pathways in endothelial and mesangial cells results in exquisite control of the process of physiological angiogenesis, with a central role played by vascular endothelial growth factor receptor 2 (VEGFR-2) and its cognate ligands. However, deregulated angiogenesis participates in numerous pathological processes. Excessive activation of VEGFR-2 has been found to mediate tissue-damaging vascular changes as well as the induction of blood vessel expansion to support the growth of solid tumors. Consequently, therapeutic intervention aimed at inhibiting the VEGFR-2 pathway has become a mainstay of treatment in cancer and retinal diseases. In this review, we introduce the concepts of physiological and pathological angiogenesis, the crucial role played by the VEGFR-2 pathway in these processes, and the various inhibitors of its activity that have entered the clinical practice. We primarily focus on the development of ramucirumab, the antagonist monoclonal antibody (mAb) that inhibits VEGFR-2 and has recently been approved for use in patients with gastric, colorectal, and lung cancers. We examine in-depth the pre-clinical studies using DC101, the mAb to mouse VEGFR-2, which provided a conceptual foundation for the role of VEGFR-2 in physiological and pathological angiogenesis. Finally, we discuss further clinical development of ramucirumab and the future of targeting the VEGF pathway for the treatment of cancer.
Collapse
|
45
|
Huang D, Zhao C, Ju R, Kumar A, Tian G, Huang L, Zheng L, Li X, Liu L, Wang S, Ren X, Ye Z, Chen W, Xing L, Chen Q, Gao Z, Mi J, Tang Z, Wang B, Zhang S, Lee C, Li X. VEGF-B inhibits hyperglycemia- and Macugen-induced retinal apoptosis. Sci Rep 2016; 6:26059. [PMID: 27189805 PMCID: PMC4870690 DOI: 10.1038/srep26059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/25/2016] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago. However, its role in hyperglycemia- and VEGF-A inhibition-induced retinal apoptosis remains unknown thus far. Yet, drugs that can block VEGF-B are being used to treat patients with diabetic retinopathy and other ocular neovascular diseases. It is therefore urgent to have a better understanding of the function of VEGF-B in these pathologies. Here, we report that both streptozotocin (STZ)-induced diabetes in rats and Macugen intravitreal injection in mice leads to retinal apoptosis in retinal ganglion cell and outer nuclear layers respectively. Importantly, VEGF-B treatment by intravitreal injection markedly reduced retinal apoptosis in both models. We further reveal that VEGF-B and its receptors, vascular endothelial growth factor 1 (VEGFR1) and neuropilin 1 (NP1), are abundantly expressed in rat retinae and choroids and are upregulated by high glucose with concomitant activation of Akt and Erk. These data highlight an important function of VEGF-B in protecting retinal cells from apoptosis induced by hyperglycemia and VEGF-A inhibition. VEGF-B may therefore have a therapeutic potential in treating various retinal degenerative diseases, and modulation of VEGF-B activity in the eye needs careful consideration.
Collapse
Affiliation(s)
- Delong Huang
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Chen Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Geng Tian
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Lei Zheng
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Xianglin Li
- Medical Imaging Institute, Shandong Province Characteristical Key Subject, Medical Imaging and Nuclear Medicine, Binzhou Medical University, Yantai, 264003 P. R. China
| | - Lixian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Zhimin Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Liying Xing
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Qishan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Zhiqin Gao
- Department of Cell Biology, Weifang Medical University, Weifang, 261053 P. R. China
| | - Jia Mi
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China
| | - Zhongshu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Bin Wang
- Medical Imaging Institute, Shandong Province Characteristical Key Subject, Medical Imaging and Nuclear Medicine, Binzhou Medical University, Yantai, 264003 P. R. China
| | - Shuping Zhang
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Xuri Li
- Center for Medical and Pharmaceutical Research, Binzhou Medical University, Yantai, Shandong, 264003, P. R. China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| |
Collapse
|
46
|
Yang Y, Andersson P, Hosaka K, Zhang Y, Cao R, Iwamoto H, Yang X, Nakamura M, Wang J, Zhuang R, Morikawa H, Xue Y, Braun H, Beyaert R, Samani N, Nakae S, Hams E, Dissing S, Fallon PG, Langer R, Cao Y. The PDGF-BB-SOX7 axis-modulated IL-33 in pericytes and stromal cells promotes metastasis through tumour-associated macrophages. Nat Commun 2016; 7:11385. [PMID: 27150562 PMCID: PMC4859070 DOI: 10.1038/ncomms11385] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/21/2016] [Indexed: 12/16/2022] Open
Abstract
Signalling molecules and pathways that mediate crosstalk between various tumour cellular compartments in cancer metastasis remain largely unknown. We report a mechanism of the interaction between perivascular cells and tumour-associated macrophages (TAMs) in promoting metastasis through the IL-33–ST2-dependent pathway in xenograft mouse models of cancer. IL-33 is the highest upregulated gene through activation of SOX7 transcription factor in PDGF-BB-stimulated pericytes. Gain- and loss-of-function experiments validate that IL-33 promotes metastasis through recruitment of TAMs. Pharmacological inhibition of the IL-33–ST2 signalling by a soluble ST2 significantly inhibits TAMs and metastasis. Genetic deletion of host IL-33 in mice also blocks PDGF-BB-induced TAM recruitment and metastasis. These findings shed light on the role of tumour stroma in promoting metastasis and have therapeutic implications for cancer therapy. Elevated IL-33 levels have been correlated with metastasis and poor prognosis. Here the authors show in mouse tumour xenograft models that PDGF-BB produced by tumour cells induces IL-33 via Sox7 in tumour pericytes, and IL-33 promotes metastasis through its effects on tumour-associated macrophages.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Patrik Andersson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Yin Zhang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Renhai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Hideki Iwamoto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Xiaojuan Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Masaki Nakamura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Jian Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Rujie Zhuang
- The TCM Hospital of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Hiromasa Morikawa
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Yuan Xue
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Harald Braun
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.,Unit of Molecular Signal Transduction in Inflammation, Inflammation Research Center VIB, B-9052 Ghent, Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.,Unit of Molecular Signal Transduction in Inflammation, Inflammation Research Center VIB, B-9052 Ghent, Belgium
| | - Nilesh Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Emily Hams
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Steen Dissing
- Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, 2200N Copenhagen, Denmark
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Robert Langer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden.,Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK.,Department of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
47
|
Endocrine vasculatures are preferable targets of an antitumor ineffective low dose of anti-VEGF therapy. Proc Natl Acad Sci U S A 2016; 113:4158-63. [PMID: 27035988 DOI: 10.1073/pnas.1601649113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Anti-VEGF-based antiangiogenic drugs are designed to block tumor angiogenesis for treatment of cancer patients. However, anti-VEGF drugs produce off-tumor target effects on multiple tissues and organs and cause broad adverse effects. Here, we show that vasculatures in endocrine organs were more sensitive to anti-VEGF treatment than tumor vasculatures. In thyroid, adrenal glands, and pancreatic islets, systemic treatment with low doses of an anti-VEGF neutralizing antibody caused marked vascular regression, whereas tumor vessels remained unaffected. Additionally, a low dose of VEGF blockade significantly inhibited the formation of thyroid vascular fenestrae, leaving tumor vascular structures unchanged. Along with vascular structural changes, the low dose of VEGF blockade inhibited vascular perfusion and permeability in thyroid, but not in tumors. Prolonged treatment with the low-dose VEGF blockade caused hypertension and significantly decreased circulating levels of thyroid hormone free-T3 and -T4, leading to functional impairment of thyroid. These findings show that the fenestrated microvasculatures in endocrine organs are more sensitive than tumor vasculatures in response to systemic anti-VEGF drugs. Thus, our data support the notion that clinically nonbeneficial treatments with anti-VEGF drugs could potentially cause adverse effects.
Collapse
|
48
|
Luo X, Jia R, Yao Q, Xu Y, Luo Z, Luo X, Wang N. Docosahexaenoic acid attenuates adipose tissue angiogenesis and insulin resistance in high fat diet-fed middle-aged mice via a sirt1-dependent mechanism. Mol Nutr Food Res 2016; 60:871-85. [PMID: 26750093 DOI: 10.1002/mnfr.201500714] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/14/2015] [Accepted: 12/29/2015] [Indexed: 02/07/2023]
Abstract
SCOPE Docosahexaenoic acid (DHA; C22: 6, n-3), one of PUFAs, exerts beneficial effects on inflammatory diseases, obesity and diabetes. Angiogenesis in adipose tissue has a major role in the development of obesity and its related metabolic complications. Inhibition of angiogenesis is an emerging strategy for the novel treatment for obesity. Thus, we examined the effect of DHA on angiogenesis in adipose tissues and investigated the underlying mechanisms. METHODS AND RESULTS In high-fat diet (HFD) fed middle-aged mice, DHA inhibited the macrophage-derived inflammation and angiogenesis in adipose tissues, reduced adipocyte size and body fat composition and improved insulin sensitivity. Moreover, DHA reversed the HFD-induced reduction of Sirt1 in adipose tissues. Interestingly, the effects of DHA were attenuated by lentivirus-mediated Sirt1 knockdown with increasing expression of markers of macrophage-derived inflammation and angiogenesis, associated with impaired insulin sensitivity. CONCLUSION Overall, our findings demonstrated that DHA reduced angiogenesis of adipose tissues and attenuated insulin resistance in HFD-induced obese mice via the activation of Sirt1.
Collapse
Affiliation(s)
- Xiaoqin Luo
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China.,Department of Medicine, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Ru Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China.,Department of Prosthodontics, Stomatological Hospital, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Qinyu Yao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yirui Xu
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zhenyu Luo
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Nanping Wang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
49
|
Inhibition of hypoxia-inducible factor via upregulation of von Hippel-Lindau protein induces "angiogenic switch off" in a hepatoma mouse model. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:15020. [PMID: 27119112 PMCID: PMC4782957 DOI: 10.1038/mto.2015.20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/15/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022]
Abstract
“Angiogenic switch off” is one of the ideal therapeutic concepts in the treatment of cancer. However, the specific molecules which can induce “angiogenic switch off” in tumor have not been identified yet. In this study, we focused on von Hippel-Lindau protein (pVHL) in hepatocellular carcinoma (HCC) and investigated the effects of sulfoquinovosyl-acylpropanediol (SQAP), a novel synthetic sulfoglycolipid, for HCC. We examined mutation ratio of VHL gene in HCC using 30 HCC samples and we treated the HCC-implanted mice with SQAP. Thirty clinical samples showed no VHL genetic mutation in HCC. SQAP significantly inhibited tumor growth by inhibiting angiogenesis in a hepatoma mouse model. SQAP induced tumor “angiogenic switch off” by decreasing hypoxia-inducible factor (HIF)-1, 2α protein via pVHL upregulation. pVHL upregulation decreased HIFα protein levels through different multiple mechanisms: (i) increasing pVHL-dependent HIFα protein degradation; (ii) decreasing HIFα synthesis with decrease of NF-κB expression; and (iii) decrease of tumor hypoxia by vascular normalization. We confirmed these antitumor effects of SQAP by the loss-of-function experiments. We found that SQAP directly bound to and inhibited transglutaminase 2. This study provides evidence that upregulation of tumor pVHL is a promising target, which can induce “angiogenic switch off” in HCC.
Collapse
|
50
|
Wang HH, Chowdhury KK, Lautt WW. A synergistic, balanced antioxidant cocktail, protects aging rats from insulin resistance and absence of meal-induced insulin sensitization (AMIS) syndrome. Molecules 2015; 20:669-82. [PMID: 25569521 PMCID: PMC6272698 DOI: 10.3390/molecules20010669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/26/2014] [Indexed: 11/16/2022] Open
Abstract
A series of in vivo and in vitro studies using animal and human models in the past 15 years have demonstrated that approximately 55% (~66% in humans) of the glucose disposal effect of an i.v. injection of insulin in the fed state is dependent on the action of a second hormone, hepatic insulin sensitizing substance (HISS), which is released from the liver and stimulates glucose uptake in muscle, heart and kidneys. Sensitization of the insulin response by a meal through release of HISS is called meal-induced insulin sensitization (MIS). Absence of HISS action results in postprandial hyperglycemia, hyperinsulinemia, hyperlipidemia, adiposity, increased free radical stress and a cluster of progressive metabolic and cardiovascular dysfunctions referred to as the AMIS (absence of meal-induced insulin sensitization) syndrome. Reduced HISS release accounts for the insulin resistance that occurs with aging and is made worse by physical inactivity and diets high in sucrose or fat. This brief review provides an update of major metabolic disturbances associated with aging due to reduction of HISS release, and the protection against these pathological changes in aging animals using a balanced synergistic antioxidant cocktail SAMEC (S-adenosylmethionine, vitamins E and C). The synergy amongst the components is consistent with the known benefits of antioxidants supplied by a mixed diet and acting through diverse mechanisms. Using only three constituents, SAMEC appears suitable as an antioxidant specifically targeting the AMIS syndrome.
Collapse
Affiliation(s)
- Hui Helen Wang
- Department of Pharmacology & Therapeutics, College of Medicine, Faculty of Health Sciences, University of Manitoba, A224-753 McDermot Avenue, Winnipeg, MB R3E 0T6, Canada.
| | - Kawshik K Chowdhury
- Department of Pharmacology & Therapeutics, College of Medicine, Faculty of Health Sciences, University of Manitoba, A224-753 McDermot Avenue, Winnipeg, MB R3E 0T6, Canada.
| | - W Wayne Lautt
- Department of Pharmacology & Therapeutics, College of Medicine, Faculty of Health Sciences, University of Manitoba, A224-753 McDermot Avenue, Winnipeg, MB R3E 0T6, Canada.
| |
Collapse
|