Elsevier

Neurobiology of Disease

Volume 68, August 2014, Pages 91-103
Neurobiology of Disease

Omega-3 polyunsaturated fatty acids enhance cerebral angiogenesis and provide long-term protection after stroke

https://doi.org/10.1016/j.nbd.2014.04.014Get rights and content

Highlights

  • n  3 PUFAs elicit long-term protection against cerebral ischemic injury.

  • n  3 PUFAs enhance post-stroke angiogenesis.

  • n  3 PUFAs promote astrocyte-mediated endothelial proliferation/barrier formation.

  • Angiopoietin 2 potentiates VEGF-induced endothelial proliferation/barrier formation.

  • VEGF-Src signaling participates in n  3 PUFA-mediated revascularization.

Abstract

Stroke is a devastating neurological disorder and one of the leading causes of death and serious disability. After cerebral ischemia, revascularization in the ischemic boundary zone provides nutritive blood flow as well as various growth factors to promote the survival and activity of neurons and neural progenitor cells. Enhancement of angiogenesis and the resulting improvement of cerebral microcirculation are key restorative mechanisms and represent an important therapeutic strategy for ischemic stroke. In the present study, we tested the hypothesis that post-stroke angiogenesis would be enhanced by omega-3 polyunsaturated fatty acids (n  3 PUFAs), a major component of dietary fish oil. To this end, we found that transgenic fat-1 mice that overproduce n  3 PUFAs exhibited long-term behavioral and histological protection against transient focal cerebral ischemia (tFCI). Importantly, fat-1 transgenic mice also exhibited robust improvements in revascularization and angiogenesis compared to wild type littermates, suggesting a potential role for n  3 fatty acids in post-stroke cerebrovascular remodeling. Mechanistically, n  3 PUFAs induced upregulation of angiopoietin 2 (Ang 2) in astrocytes after tFCI and stimulated extracellular Ang 2 release from cultured astrocytes after oxygen and glucose deprivation. Ang 2 facilitated endothelial proliferation and barrier formation in vitro by potentiating the effects of VEGF on phospholipase Cγ1 and Src signaling. Consistent with these findings, blockade of Src activity in post-stroke fat-1 mice impaired n  3 PUFA-induced angiogenesis and exacerbated long-term neurological outcomes. Taken together, our findings strongly suggest that n  3 PUFA supplementation is a potential angiogenic treatment capable of augmenting brain repair and improving long-term functional recovery after cerebral ischemia.

Introduction

Stroke is the fourth leading cause of death and the leading cause of serious long-term disability in adults in the United States (Towfighi and Saver, 2011). Intravenous thrombolysis therapy with tissue plasminogen activator (tPA), the only FDA-approved treatment for ischemic strokes, is severely limited by its short therapeutic time window of less than 4.5 h (Hacke et al., 2008). To date, neuroprotective strategies that only target injured tissue during acute stages of stroke have not been successfully translated to the clinic. In contrast, neurorestorative therapies targeting the tissue spared from direct injury have recently gained attention for their potential to enhance post-stroke brain repair processes and to promote functional recovery with a longer therapeutic time window (Zhang and Chopp, 2009).

Vascular remodeling triggered by reduction of blood flow is a major endogenous defense mechanism in the brain following stroke. Revascularization is induced by shear fluid stress and consists of arteriogenesis, or an improvement in collateral blood flow through pre-existing vasculature in the early phase after ischemic injury. In the late stages after ischemic injury, revascularization also consists of a surge in angiogenesis through endothelial cell proliferation and the subsequent formation of new blood vessels (Liu et al., 2014). Post-stroke angiogenesis greatly improves tissue perfusion and endothelial cells release a plethora of neurotrophic factors, supporting the activity of neurons and neural progenitor cells and promoting long-term functional recovery (Zhang and Chopp, 2009). Thus, post-stroke angiogenesis is an important target for therapeutic interventions. Numerous experiments have attempted to enhance post-stroke angiogenesis, such as the transplantation of endothelial progenitor cells (EPCs), which are of great therapeutic value and already under clinical investigation (Fan et al., 2010, Ishikawa et al., 2013, Liu et al., 2014, Yin et al., 2013).

Omega-3 polyunsaturated fatty acids (n  3 PUFAs) are known to protect against ischemic brain injury in several stroke models (Belayev et al., 2009, Hu et al., 2013, Zhang et al., 2010). However, the underlying mechanisms are not fully understood. Multiple mechanisms have been proposed for n  3 PUFA-mediated protection, including reduction of oxidative stress (Bazan, 2005), anti-inflammatory effects (Musiek et al., 2008, Zhang et al., 2010), induction of heme oxygenase 1 by nuclear factor E2-related factor 2 (Zhang et al., 2014), and potentiation of neurogenesis and oligodendrogenesis (Hu et al., 2013). As neurogenesis is thought to only occur within an angiogenic microenvironment (Palmer et al., 2000), it seems likely that n  3 PUFAs also promote the formation of new blood vessels. However, the effect of n  3 PUFAs on vascular remodeling after acute ischemic stroke remains to be explored.

In order to fill this gap in the field, the present study examined the impact of stroke in transgenic (Tg) mice expressing the C. elegans fat-1 gene, which encodes an n  3 fatty acid desaturase. We demonstrated that n  3 PUFA levels were elevated in these mice through increased conversion from their endogenous n  6 forms. Overproduction n  3 PUFAs in fat-1 Tg mice provided remarkable protection against focal cerebral ischemia compared to wild type littermates. Interestingly, endogenous post-stroke angiogenesis was robustly enhanced by n  3 PUFAs, in a process involving angiopoietin 2 (Ang 2) and vascular endothelial growth factor (VEGF) signaling. Our results strongly support the view that n  3 PUFA supplementation is a potential prophylactic treatment to improve tissue repair and enhance long-term functional recovery after stroke.

Section snippets

Animals

A transgenic mouse line expressing the C. elegans fat-1 gene was created as described previously (Kang et al., 2004). The fat-1 gene encodes an n  3 fatty acid desaturase that adds an extra n  3 double bond to n  6 fatty acids, thereby converting n  6 PUFAs to their corresponding n  3 forms. The coding region of C. elegans fat-1 gene was optimized for expression in mammalian cells (Wei et al., 2010), and the resultant fat-1 cDNA was driven by a cytomegalovirus (CMV) enhancer and a chicken β-actin

Transgenic overproduction of n  3 PUFAs improves neurological functions and confers long-term protection against cerebral ischemia

Expression of the fat-1 transgene resulted in a shift in the total lipid profiles from n  6 to n  3 PUFAs in all the tissues tested (tail, liver, and brain; Fig. S2A). The concentrations of three major n  3 PUFAs, eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) were significantly elevated in the brains of fat-1 transgenic (Tg) mice (Fig. S2C). To examine the effects of n  3 PUFA overproduction on ischemic brain injury, adult male fat-1 mice and their Wt

Discussion

The present report is the first mechanistic study of the role of n  3 PUFAs in post-stroke revascularization and angiogenesis. The main findings include the following: 1) transgenic overproduction of n  3 PUFAs improved post-stroke revascularization and enhanced endogenous angiogenesis; 2) n  3 PUFAs induced Ang 2 production in astrocytes, which subsequently promoted EC proliferation and barrier formation; 3) Ang 2 potentiated VEGF-mediated angiogenic effects through the downstream molecules PLCγ1

Conclusions

In summary, the present study reveals novel neurorestorative mechanisms underlying the long-term protective effects of n  3 PUFAs against cerebral ischemia. Endogenous post-stroke angiogenesis was promoted by n  3 PUFAs and the effect depended on the neurovascular niche. Our findings indicate that n  3 PUFA supplementation is a potential angiogenic treatment to enhance endogenous tissue repair and improve long-term functional recovery after stroke.

The following are the supplementary data related

Conflict of interest

None.

Acknowledgments

This project was supported by NIH grants NS036736, NS045048, and NS056118 (to J.C.), the Research Career Scientist Award from Department of Veterans Affairs and the VA RR & D Merit Review (to J.C.). L.C. was supported by the High Level Talent Fund of the Beijing Healthcare System (Grant No. 2011-3-093) and the Program for New Century Excellent Talents in University (Grant No. NCET-12-0612). J.W. was supported by the National Natural Science Foundation of China (Grant No. 81301066) and Beijing

References (56)

  • J. Turgeon et al.

    Fish oil-enriched diet protects against ischemia by improving angiogenesis, endothelial progenitor cell function and postnatal neovascularization

    Atherosclerosis

    (2013)
  • L. Zan et al.

    Src regulates angiogenic factors and vascular permeability after focal cerebral ischemia–reperfusion

    Neuroscience

    (2014)
  • Z.G. Zhang et al.

    Neurorestorative therapies for stroke: underlying mechanisms and translation to the clinic

    Lancet Neurol.

    (2009)
  • H.G. Augustin et al.

    Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system

    Nat. Rev. Mol. Cell Biol.

    (2009)
  • N.G. Bazan

    Neuroprotectin D1 (NPD1): a DHA-derived mediator that protects brain and retina against cell injury-induced oxidative stress

    Brain Pathol.

    (2005)
  • L. Belayev et al.

    Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia

    Stroke

    (2009)
  • L. Bozoyan et al.

    Astrocytes control the development of the migration-promoting vasculature scaffold in the postnatal brain via VEGF signaling

    J. Neurosci.

    (2012)
  • G. Cao et al.

    Intracellular Bax translocation after transient cerebral ischemia: implications for a role of the mitochondrial apoptotic signaling pathway in ischemic neuronal death

    J. Cereb. Blood Flow Metab.

    (2001)
  • K.M. Connor et al.

    Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis

    Nat. Med.

    (2007)
  • C. Daly et al.

    Angiopoietin-2 functions as a Tie2 agonist in tumor models, where it limits the effects of VEGF inhibition

    Cancer Res.

    (2013)
  • E. Dejana et al.

    The role of adherens junctions and VE-cadherin in the control of vascular permeability

    J. Cell Sci.

    (2008)
  • O.B. Dimitrijevic et al.

    Effects of the chemokine CCL2 on blood–brain barrier permeability during ischemia–reperfusion injury

    J. Cereb. Blood Flow Metab.

    (2006)
  • T.N. Eady et al.

    Docosahexaenoic acid signaling modulates cell survival in experimental ischemic stroke penumbra and initiates long-term repair in young and aged rats

    PLoS One

    (2012)
  • Y. Fan et al.

    Endothelial progenitor cell transplantation improves long-term stroke outcome in mice

    Ann. Neurol.

    (2010)
  • M. Fisher et al.

    Update of the stroke therapy academic industry roundtable preclinical recommendations

    Stroke

    (2009)
  • P.L. Fox et al.

    Fish oils inhibit endothelial cell production of platelet-derived growth factor-like protein

    Science

    (1988)
  • Y. Gan et al.

    Transgenic overexpression of peroxiredoxin-2 attenuates ischemic neuronal injury via suppression of a redox-sensitive pro-death signaling pathway

    Antioxid. Redox Signal.

    (2012)
  • W. Hacke et al.

    Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke

    N. Engl. J. Med.

    (2008)
  • Cited by (0)

    1

    J.W., Y.S. and L.Z. contributed equally to this work.

    View full text