Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33853948

RESUMEN

Primary Open Angle Glaucoma (POAG) is the most common form of glaucoma that leads to irreversible vision loss. Dysfunction of trabecular meshwork (TM) tissue, a major regulator of aqueous humor (AH) outflow resistance, is associated with intraocular pressure (IOP) elevation in POAG. However, the underlying pathological mechanisms of TM dysfunction in POAG remain elusive. In this regard, transient receptor potential vanilloid 4 (TRPV4) cation channels are known to be important Ca2+ entry pathways in multiple cell types. Here, we provide direct evidence supporting Ca2+ entry through TRPV4 channels in human TM cells and show that TRPV4 channels in TM cells can be activated by increased fluid flow/shear stress. TM-specific TRPV4 channel knockout in mice elevated IOP, supporting a crucial role for TRPV4 channels in IOP regulation. Pharmacological activation of TRPV4 channels in mouse eyes also improved AH outflow facility and lowered IOP. Importantly, TRPV4 channels activated endothelial nitric oxide synthase (eNOS) in TM cells, and loss of eNOS abrogated TRPV4-induced lowering of IOP. Remarkably, TRPV4-eNOS signaling was significantly more pronounced in TM cells compared to Schlemm's canal cells. Furthermore, glaucomatous human TM cells show impaired activity of TRPV4 channels and disrupted TRPV4-eNOS signaling. Flow/shear stress activation of TRPV4 channels and subsequent NO release were also impaired in glaucomatous primary human TM cells. Together, our studies demonstrate a central role for TRPV4-eNOS signaling in IOP regulation. Our results also provide evidence that impaired TRPV4 channel activity in TM cells contributes to TM dysfunction and elevated IOP in glaucoma.


Asunto(s)
Glaucoma de Ángulo Abierto/fisiopatología , Canales Catiónicos TRPV/metabolismo , Animales , Humor Acuoso/fisiología , Canales de Calcio/metabolismo , Femenino , Glaucoma/metabolismo , Glaucoma/fisiopatología , Glaucoma de Ángulo Abierto/metabolismo , Humanos , Presión Intraocular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Esclerótica/metabolismo , Transducción de Señal/fisiología , Canales Catiónicos TRPV/fisiología , Malla Trabecular/fisiología
2.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33879616

RESUMEN

Recent studies have focused on the contribution of capillary endothelial TRPV4 channels to pulmonary pathologies, including lung edema and lung injury. However, in pulmonary hypertension (PH), small pulmonary arteries are the focus of the pathology, and endothelial TRPV4 channels in this crucial anatomy remain unexplored in PH. Here, we provide evidence that TRPV4 channels in endothelial cell caveolae maintain a low pulmonary arterial pressure under normal conditions. Moreover, the activity of caveolar TRPV4 channels is impaired in pulmonary arteries from mouse models of PH and PH patients. In PH, up-regulation of iNOS and NOX1 enzymes at endothelial cell caveolae results in the formation of the oxidant molecule peroxynitrite. Peroxynitrite, in turn, targets the structural protein caveolin-1 to reduce the activity of TRPV4 channels. These results suggest that endothelial caveolin-1-TRPV4 channel signaling lowers pulmonary arterial pressure, and impairment of endothelial caveolin-1-TRPV4 channel signaling contributes to elevated pulmonary arterial pressure in PH. Thus, inhibiting NOX1 or iNOS activity, or lowering endothelial peroxynitrite levels, may represent strategies for restoring vasodilation and pulmonary arterial pressure in PH.


Asunto(s)
Caveolas/metabolismo , Endotelio Vascular/metabolismo , Ácido Peroxinitroso/metabolismo , Hipertensión Arterial Pulmonar/etiología , Canales Catiónicos TRPV/metabolismo , Animales , Presión Arterial , Humanos , Ratones Noqueados , NADPH Oxidasa 1/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteína Quinasa C/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Canales Catiónicos TRPV/genética
3.
Int J Mol Sci ; 23(13)2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35805889

RESUMEN

Glaucoma is a multifactorial disease leading to irreversible blindness. Primary open-angle glaucoma (POAG) is the most common form and is associated with the elevation of intraocular pressure (IOP). Reduced aqueous humor (AH) outflow due to trabecular meshwork (TM) dysfunction is responsible for IOP elevation in POAG. Extracellular matrix (ECM) accumulation, actin cytoskeletal reorganization, and stiffening of the TM are associated with increased outflow resistance. Transforming growth factor (TGF) ß2, a profibrotic cytokine, is known to play an important role in the development of ocular hypertension (OHT) in POAG. An appropriate mouse model is critical in understanding the underlying molecular mechanism of TGFß2-induced OHT. To achieve this, TM can be targeted with recombinant viral vectors to express a gene of interest. Lentiviruses (LV) are known for their tropism towards TM with stable transgene expression and low immunogenicity. We, therefore, developed a novel mouse model of IOP elevation using LV gene transfer of active human TGFß2 in the TM. We developed an LV vector-encoding active hTGFß2C226,228S under the control of a cytomegalovirus (CMV) promoter. Adult C57BL/6J mice were injected intravitreally with LV expressing null or hTGFß2C226,228S. We observed a significant increase in IOP 3 weeks post-injection compared to control eyes with an average delta change of 3.3 mmHg. IOP stayed elevated up to 7 weeks post-injection, which correlated with a significant drop in the AH outflow facility (40.36%). Increased expression of active TGFß2 was observed in both AH and anterior segment samples of injected mice. The morphological assessment of the mouse TM region via hematoxylin and eosin (H&E) staining and direct ophthalmoscopy examination revealed no visible signs of inflammation or other ocular abnormalities in the injected eyes. Furthermore, transduction of primary human TM cells with LV_hTGFß2C226,228S exhibited alterations in actin cytoskeleton structures, including the formation of F-actin stress fibers and crossed-linked actin networks (CLANs), which are signature arrangements of actin cytoskeleton observed in the stiffer fibrotic-like TM. Our study demonstrated a mouse model of sustained IOP elevation via lentiviral gene delivery of active hTGFß2C226,228S that induces TM dysfunction and outflow resistance.


Asunto(s)
Glaucoma de Ángulo Abierto , Hipertensión Ocular , Actinas/metabolismo , Animales , Humor Acuoso/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/metabolismo , Presión Intraocular , Ratones , Ratones Endogámicos C57BL , Hipertensión Ocular/genética , Hipertensión Ocular/metabolismo , Malla Trabecular/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo
4.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34576258

RESUMEN

Ocular hypertension (OHT) is a serious adverse effect of the widely prescribed glucocorticoid (GC) therapy and, if left undiagnosed, it can lead to glaucoma and complete blindness. Previously, we have shown that the small chemical chaperone, sodium-4-phenylbutyrate (PBA), rescues GC-induced OHT by reducing ocular endoplasmic reticulum (ER) stress. However, the exact mechanism of how PBA rescues GC-induced OHT is not completely understood. The trabecular meshwork (TM) is a filter-like specialized contractile tissue consisting of TM cells embedded within extracellular matrix (ECM) that controls intraocular pressure (IOP) by constantly regulating aqueous humor (AH) outflow. Induction of abnormal ECM deposition in TM is a hallmark of GC-induced OHT. Here, we investigated whether PBA reduces GC-induced OHT by degrading abnormal ECM deposition in TM using mouse model of GC-induced OHT, ex vivo cultured human TM tissues and primary human TM cells. We show that topical ocular eye drops of PBA (1%) significantly lowers elevated IOP in mouse model of GC-induced OHT. Importantly, PBA prevents synthesis and deposition of GC-induced ECM in TM. We report for the first time that PBA can degrade existing abnormal ECM in normal human TM cells/tissues by inducing matrix metalloproteinase (MMP)9 expression and activity. Furthermore, inhibition of MMPs activity by chemical-inhibitor (minocycline) abrogated PBA's effect on ECM reduction and its associated ER stress. Our study indicates a non-chaperone activity of PBA via activation of MMP9 that degrades abnormal ECM accumulation in TM.


Asunto(s)
Matriz Extracelular/metabolismo , Oftalmopatías/tratamiento farmacológico , Metaloproteinasa 9 de la Matriz/metabolismo , Hipertensión Ocular/tratamiento farmacológico , Fenilbutiratos/farmacología , Animales , Humor Acuoso/metabolismo , Córnea/patología , Estrés del Retículo Endoplásmico , Activación Enzimática , Fibronectinas/química , Fibronectinas/metabolismo , Glaucoma/metabolismo , Glaucoma de Ángulo Abierto/metabolismo , Humanos , Presión Intraocular , Masculino , Ratones , Ratones Endogámicos C57BL , Minociclina/farmacología , Sodio/química , Malla Trabecular
5.
J Physiol ; 598(17): 3577-3596, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32463112

RESUMEN

KEY POINTS: Endothelial cell TRPV4 (TRPV4EC ) channels exert a dilatory effect on the resting diameter of resistance mesenteric and pulmonary arteries. Functional intermediate- and small-conductance K+ (IK and SK) channels and endothelial nitric oxide synthase (eNOS) are present in the endothelium of mesenteric and pulmonary arteries. TRPV4EC sparklets preferentially couple with IK/SK channels in mesenteric arteries and with eNOS in pulmonary arteries. TRPV4EC channels co-localize with IK/SK channels in mesenteric arteries but not in pulmonary arteries, which may explain TRPV4EC -IK/SK channel coupling in mesenteric arteries and its absence in pulmonary arteries. The presence of the nitric oxide-scavenging protein, haemoglobin α, limits TRPV4EC -eNOS signalling in mesenteric arteries. Spatial proximity of TRPV4EC channels with eNOS and the absence of haemoglobin α favour TRPV4EC -eNOS signalling in pulmonary arteries. ABSTRACT: Spatially localized Ca2+ signals activate Ca2+ -sensitive intermediate- and small-conductance K+ (IK and SK) channels in some vascular beds and endothelial nitric oxide synthase (eNOS) in others. The present study aimed to uncover the signalling organization that determines selective Ca2+ signal to vasodilatory target coupling in the endothelium. Resistance-sized mesenteric arteries (MAs) and pulmonary arteries (PAs) were used as prototypes for arteries with predominantly IK/SK channel- and eNOS-dependent vasodilatation, respectively. Ca2+ influx signals through endothelial transient receptor potential vanilloid 4 (TRPV4EC ) channels played an important role in controlling the baseline diameter of both MAs and PAs. TRPV4EC channel activity was similar in MAs and PAs. However, the TRPV4 channel agonist GSK1016790A (10 nm) selectively activated IK/SK channels in MAs and eNOS in PAs, revealing preferential TRPV4EC -IK/SK channel coupling in MAs and TRPV4EC -eNOS coupling in PAs. IK/SK channels co-localized with TRPV4EC channels at myoendothelial projections (MEPs) in MAs, although they lacked the spatial proximity necessary for their activation by TRPV4EC channels in PAs. Additionally, the presence of the NO scavenging protein haemoglobin α (Hbα) within nanometer proximity to eNOS limits TRPV4EC -eNOS signalling in MAs. By contrast, co-localization of TRPV4EC channels and eNOS at MEPs, and the absence of Hbα, favour TRPV4EC -eNOS coupling in PAs. Thus, our results reveal that differential spatial organization of signalling elements determines TRPV4EC -IK/SK vs. TRPV4EC -eNOS coupling in resistance arteries.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III , Arteria Pulmonar , Endotelio Vascular , Arterias Mesentéricas , Vasodilatación
6.
Mol Vis ; 26: 434-444, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32565671

RESUMEN

Chronic elevation of intraocular pressure (IOP) is a major risk factor associated with primary open angle glaucoma (POAG), a common form of progressive optic neuropathy that can lead to debilitating loss of vision. Recent studies have identified the role of nitric oxide (NO) in the regulation of IOP, and as a result, several therapeutic ventures are currently targeting enhancement of NO signaling in the eye. Although a low level of NO is important for ocular physiology, excess exogenous NO can be detrimental. Therefore, the ability to directly measure NO in real time is essential for determining the role of NO signaling in glaucomatous pathophysiology. Historically, NO activity in human tissues has been determined by indirect methods that measure levels of NO metabolites (nitrate/nitrite) or downstream components of the NO signaling pathway (cGMP). In this proof-of-concept work, we assess the feasibility of direct, real-time measurement of NO in ex vivo cultured human corneoscleral segments using electrochemistry. A NO-selective electrode (ISO-NOPF200) paired to a free radical analyzer (TBR1025) was placed on the trabecular meshwork (TM) rim for real-time measurement of NO released from cells. Exogenous NO produced within cells was measured after treatment of corneoscleral segments with esterase-dependent NO-donor O2-acetoxymethylated diazeniumdiolate (DETA-NONOate/AM; 20 µM) and latanoprostene bunod (5-20 µM). A fluorescent NO-binding dye DAF-FM (4-Amino-5-methylamino- 2',7'-difluorofluorescein diacetate) was used for validation. A linear relationship was observed between the electric currents measured by the NO-sensing electrode and the NO standard concentrations, establishing a robust calibration curve. Treatment of ex vivo cultured human donor corneoscleral segments with DETA-NONOate/AM and latanoprostene bunod led to a significant increase in NO production compared with vehicle-treated controls, as detected electrochemically. Furthermore, the DAF-FM fluorescence intensity was higher in outflow pathway tissues of corneoscleral segments treated with DETA-NONOate/AM and latanoprostene bunod compared with vehicle-treated controls. In conclusion, these results demonstrate that NO-sensing electrodes can be used to directly measure NO levels in real time from the tissues of the outflow pathway.


Asunto(s)
Electroquímica/instrumentación , Electroquímica/métodos , Limbo de la Córnea/metabolismo , Óxido Nítrico/metabolismo , Hipertensión Ocular/metabolismo , Tonometría Ocular/instrumentación , Tonometría Ocular/métodos , Compuestos Azo/química , Células Cultivadas , Electrodos , Fluoresceínas/química , Humanos , Presión Intraocular , Limbo de la Córnea/citología , Prostaglandinas F Sintéticas/química , Malla Trabecular/metabolismo
7.
J Biol Chem ; 293(25): 9854-9868, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29743238

RESUMEN

Elevation of intraocular pressure (IOP) is a serious adverse effect of glucocorticoid (GC) therapy. Increased extracellular matrix (ECM) accumulation and endoplasmic reticulum (ER) stress in the trabecular meshwork (TM) is associated with GC-induced IOP elevation. However, the molecular mechanisms by which GCs induce ECM accumulation and ER stress in the TM have not been determined. Here, we show that a potent GC, dexamethasone (Dex), activates transforming growth factor ß (TGFß) signaling, leading to GC-induced ECM accumulation, ER stress, and IOP elevation. Dex increased both the precursor and bioactive forms of TGFß2 in conditioned medium and activated TGFß-induced SMAD signaling in primary human TM cells. Dex also activated TGFß2 in the aqueous humor and TM of a mouse model of Dex-induced ocular hypertension. We further show that Smad3-/- mice are protected from Dex-induced ocular hypertension, ER stress, and ECM accumulation. Moreover, treating WT mice with a selective TGFß receptor kinase I inhibitor, LY364947, significantly decreased Dex-induced ocular hypertension. Of note, knockdown of the ER stress-induced activating transcription factor 4 (ATF4), or C/EBP homologous protein (CHOP), completely prevented Dex-induced TGFß2 activation and ECM accumulation in TM cells. These observations suggested that chronic ER stress promotes Dex-induced ocular hypertension via TGFß signaling. Our results indicate that TGFß2 signaling plays a central role in GC-induced ocular hypertension and provides therapeutic targets for GC-induced ocular hypertension.


Asunto(s)
Dexametasona/toxicidad , Glucocorticoides/toxicidad , Hipertensión Ocular/patología , Proteína smad3/fisiología , Malla Trabecular/patología , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipertensión Ocular/inducido químicamente , Hipertensión Ocular/metabolismo , Malla Trabecular/efectos de los fármacos , Factor de Crecimiento Transformador beta2/genética
8.
Am J Pathol ; 187(4): 713-723, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28167045

RESUMEN

Glucocorticoid (GC)-induced ocular hypertension (OHT) is a serious adverse effect of prolonged GC therapy that can lead to iatrogenic glaucoma and permanent vision loss. An appropriate mouse model can help us understand precise molecular mechanisms and etiology of GC-induced OHT. We therefore developed a novel, simple, and reproducible mouse model of GC-induced OHT. GC-induced myocilin expression in the trabecular meshwork (TM) has been suggested to play an important role in GC-induced OHT. We further determined whether myocilin contributes to GC-OHT. C57BL/6J mice received weekly periocular conjunctival fornix injections of a dexamethasone-21-acetate (DEX-Ac) formulation. Intraocular pressure (IOP) elevation was relatively rapid and significant, and correlated with reduced conventional outflow facility. Nighttime IOPs were higher in ocular hypertensive eyes compared to daytime IOPs. DEX-Ac treatment led to increased expression of fibronectin, collagen I, and α-smooth muscle actin in the TM in mouse eyes. No changes in body weight indicated no systemic toxicity associated with DEX-Ac treatment. Wild-type mice showed increased myocilin expression in the TM on DEX-Ac treatment. Both wild-type and Myoc-/- mice had equivalent and significantly elevated IOP with DEX-Ac treatment every week. In conclusion, our mouse model mimics many aspects of GC-induced OHT in humans, and we further demonstrate that myocilin does not play a major role in DEX-induced OHT in mice.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Dexametasona/análogos & derivados , Proteínas del Ojo/metabolismo , Glicoproteínas/metabolismo , Hipertensión Ocular/inducido químicamente , Anestesia , Animales , Peso Corporal/efectos de los fármacos , Colágeno Tipo I/metabolismo , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Vías de Administración de Medicamentos , Esquema de Medicación , Femenino , Fibronectinas/metabolismo , Inyecciones , Inyecciones Intraoculares , Presión Intraocular , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Hipertensión Ocular/fisiopatología , Malla Trabecular/efectos de los fármacos , Malla Trabecular/patología
9.
Glia ; 62(9): 1486-501, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24839011

RESUMEN

As adenosine 5'-triphosphate (ATP) released from astrocytes can modulate many neural signaling systems, the triggers and pathways for this ATP release are important. Here, the ability of mechanical strain to trigger ATP release through pannexin channels and the effects of sustained strain on pannexin expression were examined in rat optic nerve head astrocytes. Astrocytes released ATP when subjected to 5% of equibiaxial strain or to hypotonic swelling. Although astrocytes expressed mRNA for pannexins 1-3, connexin 43, and VNUT, pharmacological analysis suggested a predominant role for pannexins in mechanosensitive ATP release, with Rho kinase contribution. Astrocytes from panx1(-/-) mice had reduced baseline and stimulated levels of extracellular ATP, confirming the role for pannexins. Swelling astrocytes triggered a regulatory volume decrease that was inhibited by apyrase or probenecid. The swelling-induced rise in calcium was inhibited by P2X7 receptor antagonists A438079 and AZ10606120, in addition to apyrase and carbenoxolone. Extended stretch of astrocytes in vitro upregulated expression of panx1 and panx2 mRNA. A similar upregulation was observed in vivo in optic nerve head tissue from the Tg-MYOC(Y437H) mouse model of chronic glaucoma; genes for panx1, panx2, and panx3 were increased, whereas immunohistochemistry confirmed increased expression of pannexin 1 protein. In summary, astrocytes released ATP in response to mechanical strain, with pannexin 1 the predominant efflux pathway. Sustained strain upregulated pannexins in vitro and in vivo. Together, these findings provide a mechanism by which extracellular ATP remains elevated under chronic mechanical strain, as found in the optic nerve head of patients with glaucoma.


Asunto(s)
Adenosina Trifosfato/metabolismo , Astrocitos/fisiología , Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Disco Óptico/fisiología , Estrés Mecánico , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Conexinas/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Femenino , Glaucoma/fisiopatología , Glicoproteínas/genética , Glicoproteínas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas de Transporte de Nucleótidos/metabolismo , Disco Óptico/efectos de los fármacos , Presión Osmótica/fisiología , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas Long-Evans
10.
Sci Rep ; 14(1): 6958, 2024 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-38521856

RESUMEN

Mutations in myocilin (MYOC) are the leading known genetic cause of primary open-angle glaucoma, responsible for about 4% of all cases. Mutations in MYOC cause a gain-of-function phenotype in which mutant myocilin accumulates in the endoplasmic reticulum (ER) leading to ER stress and trabecular meshwork (TM) cell death. Therefore, knocking out myocilin at the genome level is an ideal strategy to permanently cure the disease. We have previously utilized CRISPR/Cas9 genome editing successfully to target MYOC using adenovirus 5 (Ad5). However, Ad5 is not a suitable vector for clinical use. Here, we sought to determine the efficacy of adeno-associated viruses (AAVs) and lentiviruses (LVs) to target the TM. First, we examined the TM tropism of single-stranded (ss) and self-complimentary (sc) AAV serotypes as well as LV expressing GFP via intravitreal (IVT) and intracameral (IC) injections. We observed that LV_GFP expression was more specific to the TM injected via the IVT route. IC injections of Trp-mutant scAAV2 showed a prominent expression of GFP in the TM. However, robust GFP expression was also observed in the ciliary body and retina. We next constructed lentiviral particles expressing Cas9 and guide RNA (gRNA) targeting MYOC (crMYOC) and transduction of TM cells stably expressing mutant myocilin with LV_crMYOC significantly reduced myocilin accumulation and its associated chronic ER stress. A single IVT injection of LV_crMYOC in Tg-MYOCY437H mice decreased myocilin accumulation in TM and reduced elevated IOP significantly. Together, our data indicates, LV_crMYOC targets MYOC gene editing in TM and rescues a mouse model of myocilin-associated glaucoma.


Asunto(s)
Proteínas del Citoesqueleto , Glaucoma de Ángulo Abierto , Glicoproteínas , Animales , Ratones , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/terapia , Glaucoma de Ángulo Abierto/metabolismo , Presión Intraocular/genética , Lentivirus/genética , Malla Trabecular/metabolismo
11.
Methods Mol Biol ; 2708: 77-97, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37558962

RESUMEN

Viral transduction of the mouse trabecular meshwork using a variety of transgenes associated with glaucoma generates an inducible and reproducible method for generating ocular hypertension due to increased aqueous humor outflow resistance of the conventional outflow pathway. Both adenovirus serotype 5 (Ad5) and lentiviruses have selective tropism for the mouse trabecular meshwork with intraocular injections. Accurate intraocular pressures are easily measured using a rebound tonometer, and aqueous humor outflow facilities can be measured in anesthetized live mice.


Asunto(s)
Glaucoma , Hipertensión Ocular , Ratones , Animales , Hipertensión Ocular/genética , Hipertensión Ocular/metabolismo , Presión Intraocular , Malla Trabecular/metabolismo , Humor Acuoso/metabolismo
12.
Res Sq ; 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38196579

RESUMEN

Mutations in myocilin (MYOC) are the leading known genetic cause of primary open-angle glaucoma, responsible for about 4% of all cases. Mutations in MYOC cause a gain-of-function phenotype in which mutant myocilin accumulates in the endoplasmic reticulum (ER) leading to ER stress and trabecular meshwork (TM) cell death. Therefore, knocking out myocilin at the genome level is an ideal strategy to permanently cure the disease. We have previously utilized CRISPR/Cas9 genome editing successfully to target MYOC using adenovirus 5 (Ad5). However, Ad5 is not a suitable vector for clinical use. Here, we sought to determine the efficacy of adeno-associated viruses (AAVs) and lentiviruses (LVs) to target the TM. First, we examined the TM tropism of single-stranded (ss) and self-complimentary (sc) AAV serotypes as well as LV expressing GFP via intravitreal (IVT) and intracameral (IC) injections. We observed that LV_GFP expression was more specific to the TM injected via the IVT route. IC injections of Trp-mutant scAAV2 showed a prominent expression of GFP in the TM. However, robust GFP expression was also observed in the ciliary body and retina. We next constructed lentiviral particles expressing Cas9 and guide RNA (gRNA) targeting MYOC (crMYOC) and transduction of TM cells stably expressing mutant myocilin with LV_crMYOC significantly reduced myocilin accumulation and its associated chronic ER stress. A single IVT injection of LV_crMYOC in Tg-MYOCY437H mice decreased myocilin accumulation in TM and reduced elevated IOP significantly. Together, our data indicates, LV_crMYOC targets MYOC gene editing in TM and rescues a mouse model of myocilin-associated glaucoma.

13.
Mol Vis ; 17: 1745-58, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738403

RESUMEN

PURPOSE: Transforming growth factor-ß2 (TGF-ß2) is associated with glaucomatous neuropathy, primarily via the increased synthesis and secretion of extracellular matrix (ECM) proteins and remodeling of the optic nerve head (ONH). Here, we investigated the signaling pathways used by TGF-ß2 to stimulate ECM expression by ONH astrocytes and lamina cribrosa (LC) cells. METHODS: TGF-ß2 localization and secretion was examined in human donor tissues and ONH astrocytes and LC cells. To examine TGF-ß2 signaling, ONH astrocytes and LC cells were treated with recombinant TGF-ß2, and phosphorylation of Smad and non-Smad signaling proteins were examined by western blot analyses and immunostaining. RESULTS: TGF-ß2 is significantly increased in the LC region of the ONH in glaucomatous eyes compared to age-matched normal eyes (n=4, p<0.0013). ONH astrocytes and LC cells secrete TGF-ß2, indicating that these cells may be an in vivo source of TGF-ß2 in the human ONH. In addition, treatment of ONH astrocytes and LC cells with exogenous TGF-ß2 increased ECM protein synthesis and secretion. With respect to TGF-ß2 signaling, recombinant TGF-ß2 induced phosphorylation of canonical signaling proteins Smad2/3 but did not alter phosphorylation of non-canonical signaling proteins extracellular signal-regulated kinases (ERK)1/2, p38, and c-Jun N-terminal kinases (JNK)1/2 proteins in ONH astrocytes and LC cells. Exogenous TGF-ß2 increased co-localization of pSmad2/3 with Co-Smad4 in the nucleus of ONH astrocytes and LC cells further indicating activation of the canonical Smad signaling pathway. Furthermore, inhibition of TGF-ß I receptor activity by SB431542 or inhibition of Smad3 phosphorylation by SIS3 blocked TGF-ß2 stimulated ECM expression as well as activation of downstream canonical pathway signaling molecules. Knockdown of either Smad2 or Smad3 via small interfering RNA (siRNA) reduced TGF-ß2 stimulated ECM proteins in ONH astrocytes and LC cells. CONCLUSIONS: These studies indicate that TGF-ß2 utilizes the canonical Smad signaling pathway to stimulate ECM synthesis in human ONH cells. Our studies also indicate that pSmad2/3 is required for TGF-ß2 stimulation of ECM remodeling.


Asunto(s)
Astrocitos/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Glaucoma/metabolismo , Disco Óptico/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta2 , Anciano , Anciano de 80 o más Años , Astrocitos/citología , Astrocitos/efectos de los fármacos , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen/efectos de los fármacos , Glaucoma/genética , Glaucoma/patología , Humanos , Inmunohistoquímica , Disco Óptico/citología , Disco Óptico/efectos de los fármacos , Fosforilación/efectos de los fármacos , ARN Mensajero/análisis , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Proteínas Smad/genética , Factor de Crecimiento Transformador beta2/farmacología
14.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33539326

RESUMEN

Elevation of intraocular pressure (IOP) due to trabecular meshwork (TM) damage is associated with primary open-angle glaucoma (POAG). Myocilin mutations resulting in elevated IOP are the most common genetic causes of POAG. We have previously shown that mutant myocilin accumulates in the ER and induces chronic ER stress, leading to TM damage and IOP elevation. However, it is not understood how chronic ER stress leads to TM dysfunction and loss. Here, we report that mutant myocilin activated autophagy but was functionally impaired in cultured human TM cells and in a mouse model of myocilin-associated POAG (Tg-MYOCY437H). Genetic and pharmacological inhibition of autophagy worsened mutant myocilin accumulation and exacerbated IOP elevation in Tg-MYOCY437H mice. Remarkably, impaired autophagy was associated with chronic ER stress-induced transcriptional factor CHOP. Deletion of CHOP corrected impaired autophagy, enhanced recognition and degradation of mutant myocilin by autophagy, and reduced glaucoma in Tg-MYOCY437H mice. Stimulating autophagic flux via tat-beclin 1 peptide or torin 2 promoted autophagic degradation of mutant myocilin and reduced elevated IOP in Tg-MYOCY437H mice. Our study provides an alternate treatment strategy for myocilin-associated POAG by correcting impaired autophagy in the TM.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas del Ojo/metabolismo , Glaucoma de Ángulo Abierto/metabolismo , Glicoproteínas/metabolismo , Hipertensión Ocular/metabolismo , Malla Trabecular/metabolismo , Animales , Autofagia , Células Cultivadas , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados
15.
PLoS One ; 15(8): e0238408, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32841305

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0232111.].

16.
PLoS One ; 15(6): e0232111, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32579557

RESUMEN

Glaucoma is the second leading cause of irreversible blindness worldwide. Primary open angle glaucoma (POAG), the most common form of glaucoma, is often associated with elevation of intraocular pressure (IOP) due to the dysfunction of trabecular meshwork (TM) tissues. Currently, an ex vivo human anterior segment perfusion cultured system is widely used to study the effects of glaucoma factors and disease modifying drugs on physiological parameters like aqueous humor (AH) dynamics and IOP homeostasis. This system requires the use of freshly enucleated intact human eyes, which are sparsely available at very high cost. In this study, we explored the feasibility of using human donor corneoscleral segments for modeling morphological and biochemical changes associated with POAG. Among the number of corneas donated each year, many are deemed ineligible for transplantation due to stringent acceptance criteria. These ineligible corneoscleral segments were obtained from the Lions Eye Bank, Tampa, Florida. Each human donor anterior corneoscleral segment was dissected into four equal quadrants and cultured for 7 days by treating with the glaucoma factors dexamethasone (Dex) or recombinant transforming growth factor (TGF) ß2 or transduced with lentiviral expression vectors containing wild type (WT) and mutant myocilin. Hematoxylin and Eosin (H&E) staining analysis revealed that the TM structural integrity is maintained after 7 days in culture. Increased TUNEL positive TM cells were observed in corneoscleral quadrants treated with glaucoma factors compared to their respective controls. However, these TUNEL positive cells were mainly confined to the scleral region adjacent to the TM. Treatment of corneoscleral quadrants with Dex or TGFß2 resulted in glaucomatous changes at the TM, which included increased extracellular matrix (ECM) proteins and induction of endoplasmic reticulum (ER) stress. Western blot analysis of the conditioned medium showed an increase in ECM (fibronectin and collagen IV) levels in Dex- or TGFß2-treated samples compared to control. Lentiviral transduction of quadrants resulted in expression of WT and mutant myocilin in TM tissues. Western blot analysis of conditioned medium revealed decreased secretion of mutant myocilin compared to WT myocilin. Moreover, increased ECM deposition and ER stress induction was observed in the TM of mutant myocilin transduced quadrants. Our findings suggest that the ex-vivo cultured human corneoscleral segment model is cost-effective and can be used as a pre-screening tool to study the effects of glaucoma factors and anti-glaucoma therapeutics on the TM.


Asunto(s)
Dexametasona/farmacología , Limbo de la Córnea/metabolismo , Malla Trabecular/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Glaucoma de Ángulo Abierto/patología , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Limbo de la Córnea/citología , Limbo de la Córnea/efectos de los fármacos , Malla Trabecular/metabolismo , Malla Trabecular/patología , Regulación hacia Arriba/efectos de los fármacos
17.
Mol Neurodegener ; 15(1): 48, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32854767

RESUMEN

BACKGROUND: Glaucoma is a leading neurodegenerative disease affecting over 70 million individuals worldwide. Early pathological events of axonal degeneration and retinopathy in response to elevated intraocular pressure (IOP) are limited and not well-defined due to the lack of appropriate animal models that faithfully replicate all the phenotypes of primary open angle glaucoma (POAG), the most common form of glaucoma. Glucocorticoid (GC)-induced ocular hypertension (OHT) and its associated iatrogenic open-angle glaucoma share many features with POAG. Here, we characterized a novel mouse model of GC-induced OHT for glaucomatous neurodegeneration and further explored early pathological events of axonal degeneration in response to elevated IOP. METHODS: C57BL/6 J mice were periocularly injected with either vehicle or the potent GC, dexamethasone 21-acetate (Dex) once a week for 10 weeks. Glaucoma phenotypes including IOP, outflow facility, structural and functional loss of retinal ganglion cells (RGCs), optic nerve (ON) degeneration, gliosis, and anterograde axonal transport deficits were examined at various stages of OHT. RESULTS: Prolonged treatment with Dex leads to glaucoma in mice similar to POAG patients including IOP elevation due to reduced outflow facility and dysfunction of trabecular meshwork, progressive ON degeneration and structural and functional loss of RGCs. Lowering of IOP rescued Dex-induced ON degeneration and RGC loss, suggesting that glaucomatous neurodegeneration is IOP dependent. Also, Dex-induced neurodegeneration was associated with activation of astrocytes, axonal transport deficits, ON demyelination, mitochondrial accumulation and immune cell infiltration in the optic nerve head (ONH) region. Our studies further show that ON degeneration precedes structural and functional loss of RGCs in Dex-treated mice. Axonal damage and transport deficits initiate at the ONH and progress toward the distal end of ON and target regions in the brain (i.e. superior colliculus). Most of anterograde transport was preserved during initial stages of axonal degeneration (30% loss) and complete transport deficits were only observed at the ONH during later stages of severe axonal degeneration (50% loss). CONCLUSIONS: These findings indicate that ON degeneration and transport deficits at the ONH precede RGC structural and functional loss and provide a new potential therapeutic window for rescuing neuronal loss and restoring health of damaged axons in glaucoma.


Asunto(s)
Transporte Axonal/fisiología , Glaucoma/patología , Degeneración Nerviosa/patología , Disco Óptico/patología , Células Ganglionares de la Retina/patología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL
18.
Nat Commun ; 11(1): 5594, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154371

RESUMEN

The underlying pathological mechanisms of glaucomatous trabecular meshwork (TM) damage and elevation of intraocular pressure (IOP) are poorly understood. Here, we report that the chronic endoplasmic reticulum (ER) stress-induced ATF4-CHOP-GADD34 pathway is activated in TM of human and mouse glaucoma. Expression of ATF4 in TM promotes aberrant protein synthesis and ER client protein load, leading to TM dysfunction and cell death. These events lead to IOP elevation and glaucomatous neurodegeneration. ATF4 interacts with CHOP and this interaction is essential for IOP elevation. Notably, genetic depletion or pharmacological inhibition of ATF4-CHOP-GADD34 pathway prevents TM cell death and rescues mouse models of glaucoma by reducing protein synthesis and ER client protein load in TM cells. Importantly, glaucomatous TM cells exhibit significantly increased protein synthesis along with induction of ATF4-CHOP-GADD34 pathway. These studies indicate a pathological role of ATF4-CHOP-GADD34 pathway in glaucoma and provide a possible treatment for glaucoma by targeting this pathway.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Estrés del Retículo Endoplásmico , Glaucoma de Ángulo Abierto/metabolismo , Biosíntesis de Proteínas , Factor de Transcripción Activador 4/antagonistas & inhibidores , Factor de Transcripción Activador 4/genética , Animales , Humor Acuoso/metabolismo , Muerte Celular , Células Cultivadas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Glaucoma de Ángulo Abierto/tratamiento farmacológico , Glaucoma de Ángulo Abierto/patología , Humanos , Ratones , Hipertensión Ocular/tratamiento farmacológico , Hipertensión Ocular/metabolismo , Hipertensión Ocular/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Biosíntesis de Proteínas/efectos de los fármacos , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Transducción de Señal , Malla Trabecular/efectos de los fármacos , Malla Trabecular/metabolismo , Malla Trabecular/patología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
19.
Glia ; 57(7): 755-66, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19031438

RESUMEN

The characteristic cupping of the optic nerve head (ONH) in glaucoma is associated with elevated TGF-beta2 and increased synthesis and deposition of extracellular matrix (ECM) proteins. In addition to TGF-beta2, the human ONH also expresses bone morphogenetic proteins (BMPs) and BMP receptors, which are members of the TGF-beta superfamily. We examined the potential effects of BMP4 and the BMP antagonist gremlin on TGF-beta2 induction of ECM proteins in ONH cells. BMP-4 dose dependently inhibited TGF-beta2-induced fibronectin (FN) and PAI-1 expression in ONH astrocytes and lamina cribrosa (LC) cells and also reduced TGF-beta2 stimulation of collagen I, collagen VI, and elastin. Addition of gremlin blocked this BMP-4 response, increasing cellular and secreted FN as well as PAI-1 levels in both cell types. Gremlin was expressed in ONH tissues and ONH cells, and gremlin protein levels were significantly increased in the LC region of human glaucomatous ONH tissues. Interestingly, recombinant gremlin dose dependently increased ECM protein expression in cultured ONH astrocytes and LC cells. Gremlin stimulation of ECM required activation of TGF-beta receptor and R-Smad3. TGF-beta2 increased gremlin mRNA expression and protein levels in ONH cells. Inhibition of either the type I TGF-beta receptor or Smad3 phosphorylation blocked TGF-beta2-induced gremlin expression. In conclusion, BMP4 blocked the TGF-beta2 induction of ECM proteins in ONH cells. The BMP antagonist gremlin reversed this inhibition, allowing TGF-beta2 stimulation of ECM synthesis. Increased expression of gremlin in the glaucomatous ONH may further exacerbate TGF-beta2 effects on ONH ECM metabolism by inhibiting BMP-4 antagonism of TGF-beta2 signaling. Modulation of the ECM via gremlin provides a novel therapeutic target for glaucoma.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Disco Óptico/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Anciano , Anciano de 80 o más Años , Astrocitos/metabolismo , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/farmacología , Línea Celular , Línea Celular Tumoral , Colágeno/metabolismo , Elastina/metabolismo , Fibronectinas/metabolismo , Expresión Génica , Glaucoma/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad3/metabolismo
20.
mSphere ; 4(3)2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31068433

RESUMEN

Zika virus (ZIKV) infection during pregnancy leads to devastating fetal outcomes, including neurological (microcephaly) and ocular pathologies such as retinal lesions, optic nerve abnormalities, chorioretinal atrophy, and congenital glaucoma. Only clinical case reports have linked ZIKV infection to causing glaucoma, a major blinding eye disease. In the present study, we have investigated the role of ZIKV in glaucoma pathophysiology using in vitro and in vivo experimental models. We showed that human primary trabecular meshwork (Pr. TM) cells, as well as a human GTM3 cell line, were permissive to ZIKV infection. ZIKV induced the transcription of various genes expressing pattern recognition receptors (TLR2, TLR3, and RIG-I), cytokines/chemokines (TNF-α, IL-1ß, CCL5, and CXCL10), interferons (IFN-α2, IFN-ß1, and IFN-γ), and interferon-stimulated genes (ISG15 and OAS2) in Pr. TM cells. ZIKV infection in IFNAR1-/- and wild-type (WT) mouse eyes resulted in increased intraocular pressure (IOP) and the development of chorioretinal atrophy. Anterior chamber (AC) inoculation of ZIKV caused infectivity in iridocorneal angle and TM, leading to the death of TM cells in the mouse eyes. Moreover, anterior segment tissue of infected eyes exhibited increased expression of inflammatory mediators and interferons. Furthermore, ZIKV infection in IFNAR1-/- mice resulted in retinal ganglion cell (RGC) death and loss, coinciding with optic nerve infectivity and disruption of anterograde axonal transport. Because of similarity in glaucomatous pathologies in our study and other experimental glaucoma models, ZIKV infection can be used to study infectious triggers of glaucoma, currently an understudied area of investigation.IMPORTANCE Ocular complications due to ZIKV infection remains a major public health concern because of their ability to cause visual impairment or blindness. Most of the previous studies have shown ZIKV-induced ocular pathology in the posterior segment (i.e., retina) of the eye. However, some recent clinical reports from affected countries highlighted the importance of ZIKV in affecting the anterior segment of the eye and causing congenital glaucoma. Because glaucoma is the second leading cause of blindness worldwide, it is imperative to study ZIKV infection in causing glaucoma to identify potential targets for therapeutic intervention. In this study, we discovered that ZIKV permissively infects human TM cells and evokes inflammatory responses causing trabeculitis. Using a mouse model, we demonstrated that ZIKV infection resulted in higher IOP, increased RGC loss, and optic nerve abnormalities, the classical hallmarks of glaucoma. Collectively, our study provides new insights into ocular ZIKV infection resulting in glaucomatous pathology.


Asunto(s)
Ojo/patología , Ojo/virología , Glaucoma/virología , Malla Trabecular/virología , Infección por el Virus Zika/complicaciones , Virus Zika/patogenicidad , Animales , Muerte Celular , Línea Celular , Quimiocinas/genética , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Glaucoma/fisiopatología , Humanos , Interferones/genética , Presión Intraocular , Masculino , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/virología , Malla Trabecular/patología , Transcriptoma , Infección por el Virus Zika/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA