Skip to main content
Erschienen in: Angiogenesis 2/2020

18.12.2019 | Review Paper

Role of VEGFs in metabolic disorders

verfasst von: M. di Somma, M. Vliora, E. Grillo, B. Castro, E. Dakou, W. Schaafsma, J. Vanparijs, M. Corsini, C. Ravelli, E. Sakellariou, S. Mitola

Erschienen in: Angiogenesis | Ausgabe 2/2020

Einloggen, um Zugang zu erhalten

Abstract

Obesity and metabolic disorders are important public health problems. In this review, the role of vasculature network and VEGF in the adipose tissue maintenance and supplementation is discussed. Angiogenesis is a key process implicated in regulation of tissues homeostasis. Dysregulation of new blood vessels formation may be crucial and contribute to the onset of several pathological conditions, including metabolic syndrome-associated disorders. Adipose tissue homeostasis is fine regulated by vascular network. Vessels support adipose structure. Vasculature modulates the balance between positive and negative regulator factors. In white adipose tissue, vascular endothelial growth factor (VEGF) controls the metabolic activities of adipocytes promoting the trans-differentiation from white to beige phenotype. Trans-differentiation results in an increase of energy consumption. VEGF exerts an opposite effect on brown adipose tissue, where VEGF increases oxygen supply and improves energy expenditure inducing the whitening of adipocytes.
Literatur
3.
Zurück zum Zitat Iruela-Arispe ML, Dvorak HF (1997) Angiogenesis: a dynamic balance of stimulators and inhibitors. Thromb Haemost 78(1):672–677CrossRefPubMed Iruela-Arispe ML, Dvorak HF (1997) Angiogenesis: a dynamic balance of stimulators and inhibitors. Thromb Haemost 78(1):672–677CrossRefPubMed
24.
26.
Zurück zum Zitat Waltenberger J, Claesson-Welsh L, Siegbahn A, Shibuya M, Heldin CH (1994) Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor. J Biol Chem 269(43):26988–26995PubMed Waltenberger J, Claesson-Welsh L, Siegbahn A, Shibuya M, Heldin CH (1994) Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor. J Biol Chem 269(43):26988–26995PubMed
27.
Zurück zum Zitat Soker S, Miao HQ, Nomi M, Takashima S, Klagsbrun M (2002) VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J Cell Biochem 85(2):357–368CrossRefPubMed Soker S, Miao HQ, Nomi M, Takashima S, Klagsbrun M (2002) VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J Cell Biochem 85(2):357–368CrossRefPubMed
39.
Zurück zum Zitat Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Bock BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquiere B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW (2018) Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 21(3):425–532. https://doi.org/10.1007/s10456-018-9613-x CrossRefPubMedPubMedCentral Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Bock BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquiere B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW (2018) Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 21(3):425–532. https://​doi.​org/​10.​1007/​s10456-018-9613-x CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Mitola S, Soldi R, Zanon I, Barra L, Gutierrez MI, Berkhout B, Giacca M, Bussolino F (2000) Identification of specific molecular structures of human immunodeficiency virus type 1 Tat relevant for its biological effects on vascular endothelial cells. J Virol 74(1):344–353CrossRefPubMedPubMedCentral Mitola S, Soldi R, Zanon I, Barra L, Gutierrez MI, Berkhout B, Giacca M, Bussolino F (2000) Identification of specific molecular structures of human immunodeficiency virus type 1 Tat relevant for its biological effects on vascular endothelial cells. J Virol 74(1):344–353CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Yang X, Cepko CL (1996) Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 16(19):6089–6099CrossRefPubMedPubMedCentral Yang X, Cepko CL (1996) Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 16(19):6089–6099CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Casella I, Feccia T, Chelucci C, Samoggia P, Castelli G, Guerriero R, Parolini I, Petrucci E, Pelosi E, Morsilli O, Gabbianelli M, Testa U, Peschle C (2003) Autocrine-paracrine VEGF loops potentiate the maturation of megakaryocytic precursors through Flt1 receptor. Blood 101(4):1316–1323. https://doi.org/10.1182/blood-2002-07-2184 CrossRefPubMed Casella I, Feccia T, Chelucci C, Samoggia P, Castelli G, Guerriero R, Parolini I, Petrucci E, Pelosi E, Morsilli O, Gabbianelli M, Testa U, Peschle C (2003) Autocrine-paracrine VEGF loops potentiate the maturation of megakaryocytic precursors through Flt1 receptor. Blood 101(4):1316–1323. https://​doi.​org/​10.​1182/​blood-2002-07-2184 CrossRefPubMed
47.
Zurück zum Zitat Pisacane AM, Risio M (2005) VEGF and VEGFR-2 immunohistochemistry in human melanocytic naevi and cutaneous melanomas. Melanoma Res 15(1):39–43CrossRefPubMed Pisacane AM, Risio M (2005) VEGF and VEGFR-2 immunohistochemistry in human melanocytic naevi and cutaneous melanomas. Melanoma Res 15(1):39–43CrossRefPubMed
48.
Zurück zum Zitat List AF (2001) Vascular endothelial growth factor signaling pathway as an emerging target in hematologic malignancies. Oncologist 6(Suppl 5):24–31CrossRefPubMed List AF (2001) Vascular endothelial growth factor signaling pathway as an emerging target in hematologic malignancies. Oncologist 6(Suppl 5):24–31CrossRefPubMed
50.
Zurück zum Zitat Witmer AN, Blaauwgeers HG, Weich HA, Alitalo K, Vrensen GF, Schlingemann RO (2002) Altered expression patterns of VEGF receptors in human diabetic retina and in experimental VEGF-induced retinopathy in monkey. Invest Ophthalmol Vis Sci 43(3):849–857PubMed Witmer AN, Blaauwgeers HG, Weich HA, Alitalo K, Vrensen GF, Schlingemann RO (2002) Altered expression patterns of VEGF receptors in human diabetic retina and in experimental VEGF-induced retinopathy in monkey. Invest Ophthalmol Vis Sci 43(3):849–857PubMed
53.
Zurück zum Zitat Aldhahi W, Hamdy O (2003) Adipokines, inflammation, and the endothelium in diabetes. Curr Diab Rep 3(4):293–298CrossRefPubMed Aldhahi W, Hamdy O (2003) Adipokines, inflammation, and the endothelium in diabetes. Curr Diab Rep 3(4):293–298CrossRefPubMed
60.
Zurück zum Zitat Girousse A, Gil-Ortega M, Bourlier V, Bergeaud C, Sastourne-Arrey Q, Moro C, Barreau C, Guissard C, Vion J, Arnaud E, Pradere JP, Juin N, Casteilla L, Sengenes C (2019) The release of adipose stromal cells from subcutaneous adipose tissue regulates ectopic intramuscular adipocyte deposition. Cell Rep 27 (2):323–333 e325. https://doi.org/10.1016/j.celrep.2019.03.038 CrossRefPubMed Girousse A, Gil-Ortega M, Bourlier V, Bergeaud C, Sastourne-Arrey Q, Moro C, Barreau C, Guissard C, Vion J, Arnaud E, Pradere JP, Juin N, Casteilla L, Sengenes C (2019) The release of adipose stromal cells from subcutaneous adipose tissue regulates ectopic intramuscular adipocyte deposition. Cell Rep 27 (2):323–333 e325. https://​doi.​org/​10.​1016/​j.​celrep.​2019.​03.​038 CrossRefPubMed
68.
Zurück zum Zitat Gil-Ortega M, Garidou L, Barreau C, Maumus M, Breasson L, Tavernier G, Garcia-Prieto CF, Bouloumie A, Casteilla L, Sengenes C (2013) Native adipose stromal cells egress from adipose tissue in vivo: evidence during lymph node activation. Stem Cells 31(7):1309–1320. https://doi.org/10.1002/stem.1375 CrossRefPubMed Gil-Ortega M, Garidou L, Barreau C, Maumus M, Breasson L, Tavernier G, Garcia-Prieto CF, Bouloumie A, Casteilla L, Sengenes C (2013) Native adipose stromal cells egress from adipose tissue in vivo: evidence during lymph node activation. Stem Cells 31(7):1309–1320. https://​doi.​org/​10.​1002/​stem.​1375 CrossRefPubMed
73.
Zurück zum Zitat Rennert RC, Sorkin M, Januszyk M, Duscher D, Kosaraju R, Chung MT, Lennon J, Radiya-Dixit A, Raghvendra S, Maan ZN, Hu MS, Rajadas J, Rodrigues M, Gurtner GC (2014) Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations. Stem Cell Res Therapy 5(3):79. https://doi.org/10.1186/scrt468 CrossRef Rennert RC, Sorkin M, Januszyk M, Duscher D, Kosaraju R, Chung MT, Lennon J, Radiya-Dixit A, Raghvendra S, Maan ZN, Hu MS, Rajadas J, Rodrigues M, Gurtner GC (2014) Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations. Stem Cell Res Therapy 5(3):79. https://​doi.​org/​10.​1186/​scrt468 CrossRef
80.
84.
Zurück zum Zitat Matthias A, Ohlson KB, Fredriksson JM, Jacobsson A, Nedergaard J, Cannon B (2000) Thermogenic responses in brown fat cells are fully UCP1-dependent. UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty scid-induced thermogenesis. J Biol Chem 275 (33):25073–25081. https://doi.org/10.1074/jbc.M000547200 CrossRefPubMed Matthias A, Ohlson KB, Fredriksson JM, Jacobsson A, Nedergaard J, Cannon B (2000) Thermogenic responses in brown fat cells are fully UCP1-dependent. UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty scid-induced thermogenesis. J Biol Chem 275 (33):25073–25081. https://​doi.​org/​10.​1074/​jbc.​M000547200 CrossRefPubMed
85.
Zurück zum Zitat Bouillaud F, Ricquier D, Gulik-Krzywicki T, Gary-Bobo CM (1983) The possible proton translocating activity of the mitochondrial uncoupling protein of brown adipose tissue. Reconstitution studies in liposomes. FEBS Lett 164(2):272–276CrossRefPubMed Bouillaud F, Ricquier D, Gulik-Krzywicki T, Gary-Bobo CM (1983) The possible proton translocating activity of the mitochondrial uncoupling protein of brown adipose tissue. Reconstitution studies in liposomes. FEBS Lett 164(2):272–276CrossRefPubMed
87.
Zurück zum Zitat Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass CA, Huang TL, Roberts-Toler C, Weiner LS, Sze C, Chacko AT, Deschamps LN, Herder LM, Truchan N, Glasgow AL, Holman AR, Gavrila A, Hasselgren PO, Mori MA, Molla M, Tseng YH (2013) Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat Med 19(5):635–639. https://doi.org/10.1038/nm.3112 CrossRefPubMedPubMedCentral Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass CA, Huang TL, Roberts-Toler C, Weiner LS, Sze C, Chacko AT, Deschamps LN, Herder LM, Truchan N, Glasgow AL, Holman AR, Gavrila A, Hasselgren PO, Mori MA, Molla M, Tseng YH (2013) Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat Med 19(5):635–639. https://​doi.​org/​10.​1038/​nm.​3112 CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Schulz TJ, Huang TL, Tran TT, Zhang H, Townsend KL, Shadrach JL, Cerletti M, McDougall LE, Giorgadze N, Tchkonia T, Schrier D, Falb D, Kirkland JL, Wagers AJ, Tseng YH (2011) Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat. Proc Natl Acad Sci USA 108(1):143–148. https://doi.org/10.1073/pnas.1010929108 CrossRefPubMed Schulz TJ, Huang TL, Tran TT, Zhang H, Townsend KL, Shadrach JL, Cerletti M, McDougall LE, Giorgadze N, Tchkonia T, Schrier D, Falb D, Kirkland JL, Wagers AJ, Tseng YH (2011) Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat. Proc Natl Acad Sci USA 108(1):143–148. https://​doi.​org/​10.​1073/​pnas.​1010929108 CrossRefPubMed
97.
Zurück zum Zitat Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM (1998) A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92(6):829–839CrossRefPubMed Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM (1998) A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92(6):829–839CrossRefPubMed
108.
Zurück zum Zitat Hondares E, Mora O, Yubero P, Rodriguez de la Concepcion M, Iglesias R, Giralt M, Villarroya F (2006) Thiazolidinediones and rexinoids induce peroxisome proliferator-activated receptor-coactivator (PGC)-1alpha gene transcription: an autoregulatory loop controls PGC-1alpha expression in adipocytes via peroxisome proliferator-activated receptor-gamma coactivation. Endocrinology 147(6):2829–2838. https://doi.org/10.1210/en.2006-0070 CrossRefPubMed Hondares E, Mora O, Yubero P, Rodriguez de la Concepcion M, Iglesias R, Giralt M, Villarroya F (2006) Thiazolidinediones and rexinoids induce peroxisome proliferator-activated receptor-coactivator (PGC)-1alpha gene transcription: an autoregulatory loop controls PGC-1alpha expression in adipocytes via peroxisome proliferator-activated receptor-gamma coactivation. Endocrinology 147(6):2829–2838. https://​doi.​org/​10.​1210/​en.​2006-0070 CrossRefPubMed
109.
115.
Zurück zum Zitat Hagberg CE, Falkevall A, Wang X, Larsson E, Huusko J, Nilsson I, van Meeteren LA, Samen E, Lu L, Vanwildemeersch M, Klar J, Genove G, Pietras K, Stone-Elander S, Claesson-Welsh L, Yla-Herttuala S, Lindahl P, Eriksson U (2010) Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature 464(7290):917–921. https://doi.org/10.1038/nature08945 CrossRefPubMed Hagberg CE, Falkevall A, Wang X, Larsson E, Huusko J, Nilsson I, van Meeteren LA, Samen E, Lu L, Vanwildemeersch M, Klar J, Genove G, Pietras K, Stone-Elander S, Claesson-Welsh L, Yla-Herttuala S, Lindahl P, Eriksson U (2010) Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature 464(7290):917–921. https://​doi.​org/​10.​1038/​nature08945 CrossRefPubMed
116.
Zurück zum Zitat Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach KA, Bostrom EA, Choi JH, Long JZ, Kajimura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Hojlund K, Gygi SP, Spiegelman BM (2012) A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481(7382):463–468. https://doi.org/10.1038/nature10777 CrossRefPubMedPubMedCentral Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach KA, Bostrom EA, Choi JH, Long JZ, Kajimura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Hojlund K, Gygi SP, Spiegelman BM (2012) A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481(7382):463–468. https://​doi.​org/​10.​1038/​nature10777 CrossRefPubMedPubMedCentral
Metadaten
Titel
Role of VEGFs in metabolic disorders
verfasst von
M. di Somma
M. Vliora
E. Grillo
B. Castro
E. Dakou
W. Schaafsma
J. Vanparijs
M. Corsini
C. Ravelli
E. Sakellariou
S. Mitola
Publikationsdatum
18.12.2019
Verlag
Springer Netherlands
Erschienen in
Angiogenesis / Ausgabe 2/2020
Print ISSN: 0969-6970
Elektronische ISSN: 1573-7209
DOI
https://doi.org/10.1007/s10456-019-09700-1

Weitere Artikel der Ausgabe 2/2020

Angiogenesis 2/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhöhte Mortalität bei postpartalem Brustkrebs

07.05.2024 Mammakarzinom Nachrichten

Auch für Trägerinnen von BRCA-Varianten gilt: Erkranken sie fünf bis zehn Jahre nach der letzten Schwangerschaft an Brustkrebs, ist das Sterberisiko besonders hoch.

Hypertherme Chemotherapie bietet Chance auf Blasenerhalt

07.05.2024 Harnblasenkarzinom Nachrichten

Eine hypertherme intravesikale Chemotherapie mit Mitomycin kann für Patienten mit hochriskantem nicht muskelinvasivem Blasenkrebs eine Alternative zur radikalen Zystektomie darstellen. Kölner Urologen berichten über ihre Erfahrungen.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Medizinstudium Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Vorhofflimmern bei Jüngeren gefährlicher als gedacht

06.05.2024 Vorhofflimmern Nachrichten

Immer mehr jüngere Menschen leiden unter Vorhofflimmern. Betroffene unter 65 Jahren haben viele Risikofaktoren und ein signifikant erhöhtes Sterberisiko verglichen mit Gleichaltrigen ohne die Erkrankung.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.