Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Xiaowu Gu is active.

Publication


Featured researches published by Xiaowu Gu.


American Journal of Pathology | 2014

Peroxisome Proliferator–Activated Receptor α Protects Capillary Pericytes in the Retina

Lexi Ding; Rui Cheng; Yang Hu; Yusuke Takahashi; Alicia J. Jenkins; Anthony Keech; Kenneth M. Humphries; Xiaowu Gu; Michael H. Elliott; Xiaobo Xia; Jian Xing Ma

Pericyte degeneration is an early event in diabetic retinopathy and plays an important role in progression of diabetic retinopathy. Clinical studies have shown that fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, has robust therapeutic effects on diabetic retinopathy in type 2 diabetic patients. We evaluated the protective effect of PPARα against pericyte loss in diabetic retinopathy. In streptozotocin-induced diabetic mice, fenofibrate treatment significantly ameliorated retinal acellular capillary formation and pericyte loss. In contrast, PPARα(-/-) mice with diabetes developed more severe retinal acellular capillary formation and pericyte dropout, compared with diabetic wild-type mice. Furthermore, PPARα knockout abolished the protective effect of fenofibrate against diabetes-induced retinal pericyte loss. In cultured primary human retinal capillary pericytes, activation and expression of PPARα both significantly reduced oxidative stress-induced apoptosis, decreased reactive oxygen species production, and down-regulated NAD(P)H oxidase 4 expression through blockade of NF-κB activation. Furthermore, activation and expression of PPARα both attenuated the oxidant-induced suppression of mitochondrial O2 consumption in human retinal capillary pericytes. Primary retinal pericytes from PPARα(-/-) mice displayed more apoptosis, compared with those from wild-type mice under the same oxidative stress. These findings identified a protective effect of PPARα on retinal pericytes, a novel function of endogenous PPARα in the retina.


American Journal of Pathology | 2014

Loss of Caveolin-1 Causes Blood–Retinal Barrier Breakdown, Venous Enlargement, and Mural Cell Alteration

Xiaowu Gu; Steven J. Fliesler; You Yang Zhao; William B. Stallcup; Alex Cohen; Michael H. Elliott

Blood-retinal barrier (BRB) breakdown and related vascular changes are implicated in several ocular diseases. The molecules and mechanisms regulating BRB integrity and pathophysiology are not fully elucidated. Caveolin-1 (Cav-1) ablation results in loss of caveolae and microvascular pathologies, but the role of Cav-1 in the retina is largely unknown. We examined BRB integrity and vasculature in Cav-1 knockout mice and found a significant increase in BRB permeability, compared with wild-type controls, with branch veins being frequent sites of breakdown. Vascular hyperpermeability occurred without apparent alteration in junctional proteins. Such hyperpermeability was not rescued by inhibiting eNOS activity. Veins of Cav-1 knockout retinas exhibited additional pathological features, including i) eNOS-independent enlargement, ii) altered expression of mural cell markers (eg, down-regulation of NG2 and up-regulation of αSMA), and iii) dramatic alterations in mural cell phenotype near the optic nerve head. We observed a significant NO-dependent increase in retinal artery diameter in Cav-1 knockout mice, suggesting that Cav-1 plays a role in autoregulation of resistance vessels in the retina. These findings implicate Cav-1 in maintaining BRB integrity in retinal vasculature and suggest a previously undefined role in the retinal venous system and associated mural cells. Our results are relevant to clinically significant retinal disorders with vascular pathologies, including diabetic retinopathy, uveoretinitis, and primary open-angle glaucoma.


Advances in Experimental Medicine and Biology | 2014

Spatial and Temporal Localization of Caveolin-1 Protein in the Developing Retina

Xiaowu Gu; Alaina Reagan; Allen Yen; Faizah Bhatti; Alex Cohen; Michael H. Elliott

Caveolin-1 (Cav-1), the signature protein of caveolae is expressed in several cell types in the adult retina and is linked to ocular pathologies including uveitis, diabetic retinopathy, and primary open angle glaucoma. Genetic ablation of Cav-1 causes retinal functional deficits due to disruptions in environmental homeostasis. To better understand Cav-1 function in the retina, we examined its expression/localization during postnatal retinal development. From P0-P5, Cav-1 was detected only in the developing superficial retinal vessels, in hyaloid and choroidal vasculature, and in the retinal pigment epithelium (RPE). At P7, staining began to be observed centrally in radial cells in the neuroretina, and this staining increased dramatically by P9/10 in identifiable Müller glia. Prominent vascular staining continued throughout development. These results support the idea that Cav-1 is an indicator of Müller glial differentiation and suggests that it plays an important role in Müller cell function.


Scientific Reports | 2016

Caveolin-1 modulates intraocular pressure: implications for caveolae mechanoprotection in glaucoma

Michael H. Elliott; Nicole Ashpole; Xiaowu Gu; Leonie Herrnberger; Mark E. McClellan; Gina L. Griffith; Alaina Reagan; Timothy M. Boyce; Masaki Tanito; Ernst R. Tamm; W. Daniel Stamer

Polymorphisms in the CAV1/2 genes that encode signature proteins of caveolae are associated with glaucoma, the second leading cause of blindness worldwide, and with its major risk factor, intraocular pressure (IOP). We hypothesized that caveolin-1 (Cav-1) participates in IOP maintenance via modulation of aqueous humor drainage from the eye. We localize caveolae proteins to human and murine conventional drainage tissues and show that caveolae respond to mechanical stimulation. We show that Cav-1-deficient (Cav-1−/−) mice display ocular hypertension explained by reduced pressure-dependent drainage of aqueous humor. Cav-1 deficiency results in loss of caveolae in the Schlemm’s canal (SC) and trabecular meshwork. However, their absence did not appear to impact development nor adult form of the conventional outflow tissues according to rigorous quantitative ultrastructural analyses, but did affect cell and tissue behavior. Thus, when IOP is experimentally elevated, cells of the Cav-1−/− outflow tissues are more susceptible to plasma membrane rupture indicating that caveolae play a role in mechanoprotection. Additionally, aqueous drainage from Cav-1−/− eyes was more sensitive to nitric oxide (NO) synthase inhibition than controls, suggesting that excess NO partially compensates for outflow pathway dysfunction. These results provide a functional link between a glaucoma risk gene and glaucoma-relevant pathophysiology.


Investigative Ophthalmology & Visual Science | 2014

Caveolin-1 Increases Proinflammatory Chemoattractants and Blood–Retinal Barrier Breakdown but Decreases Leukocyte Recruitment in Inflammation

Xiaoman Li; Xiaowu Gu; Timothy M. Boyce; Min Zheng; Alaina Reagan; Hui Qi; Nawajes A. Mandal; Alex Cohen; Michelle C. Callegan; Daniel J. J. Carr; Michael H. Elliott

PURPOSE Caveolin-1 (Cav-1), the signature protein of caveolae, modulates inflammatory responses, and innate immunity. However, Cav-1s role in retinal inflammation has not been rigorously tested. In this study, we examined the effect of Cav-1 ablation on the sensitivity of the retina to inflammation. METHODS Cav-1 knockout (KO) mice were challenged by intravitreal injection of lipopolysaccharide (LPS) and inflammatory cell recruitment was assessed by flow cytometry and immunohistochemistry. Leukostasis was assessed in retinal flatmounts after perfusion with FITC-labeled Concanavalin A (FITC-ConA). Chemoattractants were measured by multiplex immunoassays. Blood-retinal barrier (BRB) breakdown was assessed quantitatively by a FITC-dextran permeability assay. The ratio of extravascular to total immune cells was determined by CD45 immunohistochemistry of retinal flatmounts. RESULTS Inflammatory challenge resulted in significant blunting of proinflammatory cytokine (monocyte chemoattractant protein-1 [MCP-1/CCL2], CXCL1/KC, IL-6, and IL-1β) responses as well as reduced inflammatory BRB breakdown in Cav-1 KO retinas. Paradoxically, Cav-1 deficiency resulted in significantly increased recruitment of immune cells compared with controls as well as increased leukostasis. A similar ratio of extravascular/total leukocytes were found in Cav-1 KO and wild-type (WT) retinas suggesting that Cav-1 deficient leukocytes were as competent to extravasate as those from WT mice. We found increased levels of circulating immune cells in naïve (not challenged with LPS) Cav-1 KO mice compared with controls. CONCLUSIONS Caveolin-1 paradoxically modulates inflammatory signaling and leukocyte infiltration through distinct mechanisms. We hypothesize that Cav-1 expression may enhance inflammatory signaling while at the same time supporting the physical properties of the BRB.


Progress in Retinal and Eye Research | 2017

Caveolins and caveolae in ocular physiology and pathophysiology.

Xiaowu Gu; Alaina Reagan; Mark E. McClellan; Michael H. Elliott

ABSTRACT Caveolae are specialized, invaginated plasma membrane domains that are defined morphologically and by the expression of signature proteins called, caveolins. Caveolae and caveolins are abundant in a variety of cell types including vascular endothelium, glia, and fibroblasts where they play critical roles in transcellular transport, endocytosis, mechanotransduction, cell proliferation, membrane lipid homeostasis, and signal transduction. Given these critical cellular functions, it is surprising that ablation of the caveolae organelle does not result in lethality suggesting instead that caveolae and caveolins play modulatory roles in cellular homeostasis. Caveolar components are also expressed in ocular cell types including retinal vascular cells, Müller glia, retinal pigment epithelium (RPE), conventional aqueous humor outflow cells, the corneal epithelium and endothelium, and the lens epithelium. In the eye, studies of caveolae and other membrane microdomains (i.e., “lipid rafts”) have lagged behind what is a substantial body of literature outside vision science. However, interest in caveolae and their molecular components has increased with accumulating evidence of important roles in vision‐related functions such as blood‐retinal barrier homeostasis, ocular inflammatory signaling, pathogen entry at the ocular surface, and aqueous humor drainage. The recent association of CAV1/2 gene loci with primary open angle glaucoma and intraocular pressure has further enhanced the need to better understand caveolar functions in the context of ocular physiology and disease. Herein, we provide the first comprehensive review of literature on caveolae, caveolins, and other membrane domains in the context of visual system function. This review highlights the importance of caveolae domains and their components in ocular physiology and pathophysiology and emphasizes the need to better understand these important modulators of cellular function.


Advances in Experimental Medicine and Biology | 2016

Retinal Caveolin-1 Modulates Neuroprotective Signaling

Alaina Reagan; Xiaowu Gu; Stefanie M. Hauck; John D. Ash; Guangwen Cao; Timothy C. Thompson; Michael H. Elliott

Caveolin-1 (Cav-1), the scaffolding protein of caveolae, is expressed in several retinal cell types and is associated with ocular pathologies. Cav-1 modulates neuroinflammatory/neuroprotective responses to central nervous system injury. We have shown that loss of Cav-1 results in a blunted cytokine response in retinas challenged with inflammatory stimuli. As neuroinflammatory and neuroprotective signaling overlap in their cytokine production and downstream signaling pathways, we hypothesized that loss of Cav-1 may also suppress neuroprotective signaling in the retina. To test this, we subjected mice in which Cav-1 was deleted specifically in the retina to a neurodegenerative insult induced by sodium iodate (NaIO3) and measured STAT3 activation, a measure of neuroprotective signaling. Our results show that Cav-1 ablation blunts STAT3 activation induced by NaIO3. STAT3 activation in response to intravitreal administration of the IL-6 family cytokine, leukemia inhibitory factor (LIF), was not affected by Cav-1 deletion indicating a competent gp130 receptor response. Thus, Cav-1 modulates neuroprotective signaling by regulating the endogenous production of neuroprotective factors.


Neurobiology of Aging | 2018

Age-related focal loss of contractile vascular smooth muscle cells in retinal arterioles is accelerated by caveolin-1 deficiency

Alaina Reagan; Xiaowu Gu; Sijalu Paudel; Nicole M. Ashpole; Michelle Zalles; William E. Sonntag; Zoltan Ungvari; Anna Csiszar; Laura Otalora; Willard M. Freeman; Michael B. Stout; Michael H. Elliott

Cerebral microcirculation is critical for the preservation of brain health, and vascular impairment is associated with age-related neurodegenerative diseases. Because the retina is a component of the central nervous system, cellular changes that occur in the aging retina are likely relevant to the aging brain, and the retina provides the advantage that the entire vascular bed is visible, en face. In this study, we tested the hypothesis that normal, healthy aging alters the contractile vascular smooth muscle cell (VSMC) coverage of retinal arterioles. We found that aging results in a significant reduction of contractile VSMCs in focal patches along arterioles. Focal loss of contractile VSMCs occurs at a younger age in mice deficient in the senescence-associated protein, caveolin-1. Age-related contractile VSMC loss is not exacerbated by genetic depletion of insulin-like growth factor-1. The patchy loss of contractile VSMCs provides a cellular explanation for previous clinical studies showing focal microirregularities in retinal arteriolar responsiveness in healthy aged human subjects and is likely to contribute to age-related retinal vascular complications.


Investigative Ophthalmology & Visual Science | 2014

Caveolin-1 as a Novel Corneal Wound Healing Therapeutic Target

Gina L. Griffith; Timothy M. Boyce; Xiaowu Gu; Michael H. Elliott; Alex Cohen


Investigative Ophthalmology & Visual Science | 2014

Role of Caveolin-1 in Intraocular Pressure and Conventional Outflow Regulation

Michael H. Elliott; Xiaowu Gu; Nicole Ashpole; Gina L. Griffith; Timothy M. Boyce; Masaki Tanito; Ernst R. Tamm; W. Daniel Stamer

Collaboration


Dive into the Xiaowu Gu's collaboration.

Top Co-Authors

Avatar

Michael H. Elliott

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Alaina Reagan

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Alex Cohen

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Timothy M. Boyce

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Daniel J. J. Carr

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Gina L. Griffith

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

John D. Ash

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Nawajes A. Mandal

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Csiszar

University of Oklahoma Health Sciences Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge