Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Optom Vis Sci ; 100(11): 794-798, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37889985

ABSTRACT

SIGNIFICANCE: Stakeholder engagement has been identified by national health organizations as a crucial step to successful translation of new health care treatments. In this clinical report, clinician-stakeholder feedback is presented for the magnetic levator prosthesis (MLP), a promising noninvasive spectacle device that restores eyelid motility with magnetic force. PURPOSE: This study aimed to evaluate MLP clinical need and translational barriers. METHODS: Ten vision rehabilitation optometrists who attended an educational presentation on the MLP and participated in a hands-on workshop in the fitting of a patient were invited to complete an anonymous online survey. Ten multiple-choice items gathered data on estimated patient need, current approaches, main barriers to MLP, temporary versus chronic use, cost barriers, and need for insurance coverage. Open fields allowed for additional comments. RESULTS: Nine of 10 specialists completed the survey. Of those, seven answered that they could potentially see at least 1 to 5 patients for ptosis management within a year. The most common ptosis management options reported were the ptosis crutch, taping the eyelid open, and oxymetazoline drops, all with six responses each. Seven clinicians indicated that cost was a main concern. If cost to patient was not a barrier, all indicated they would be at least somewhat likely to try the MLP (1) for temporary management of ptosis, (2) as a pre-surgical trial, and (3) for long-term management of ptosis, with more selecting extremely likely and very likely than somewhat likely. Main comments were expressing enthusiasm for the technology and that it would be more appealing for patients if covered by insurance. CONCLUSIONS: This clinical report suggests that the main barriers to clinical success of the MLP may be cost and insurance coverage, appearance of the device, and self-application. Possible solutions are cost-benefit analysis research, engineering efforts to reduce spectacle magnet size and improve the ease of eyelid magnet application.


Subject(s)
Blepharoptosis , Humans , Feedback , Blepharoptosis/surgery , Eyelids , Prostheses and Implants , Magnetic Phenomena
2.
Proc Natl Acad Sci U S A ; 117(38): 23336-23338, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32900927

ABSTRACT

Colony-stimulating factor 1 receptor (CSF1R) inhibition has been proposed as a method for microglia depletion, with the assumption that it does not affect peripheral immune cells. Here, we show that CSF1R inhibition by PLX5622 indeed affects the myeloid and lymphoid compartments, causes long-term changes in bone marrow-derived macrophages by suppressing interleukin 1ß, CD68, and phagocytosis but not CD208, following exposure to endotoxin, and also reduces the population of resident and interstitial macrophages of peritoneum, lung, and liver but not spleen. Thus, small-molecule CSF1R inhibition is not restricted to microglia, causing strong effects on circulating and tissue macrophages that perdure long after cessation of the treatment. Given that peripheral monocytes repopulate the central nervous system after CSF1R inhibition, these changes have practical implications for relevant experimental data.


Subject(s)
Hematopoiesis/drug effects , Macrophages/drug effects , Macrophages/immunology , Microglia/drug effects , Organic Chemicals/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , Female , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Phagocytosis/drug effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Species Specificity
3.
Proc Natl Acad Sci U S A ; 116(47): 23705-23713, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685620

ABSTRACT

Inflammation plays an important role in pathological angiogenesis. Receptor-interacting protein 1 (RIP1) is highly expressed in inflammatory cells and is known to play an important role in the regulation of apoptosis, necroptosis, and inflammation; however, a comprehensive description of its role in angiogenesis remains elusive. Here, we show that RIP1 is abundantly expressed in infiltrating macrophages during angiogenesis, and genetic or pharmacological inhibition of RIP1 kinase activity using kinase-inactive RIP1K45A/K45A mice or necrostatin-1 attenuates angiogenesis in laser-induced choroidal neovascularization, Matrigel plug angiogenesis, and alkali injury-induced corneal neovascularization in mice. The inhibitory effect on angiogenesis is mediated by caspase activation through a kinase-independent function of RIP1 and RIP3. Mechanistically, infiltrating macrophages are the key target of RIP1 kinase inhibition to attenuate pathological angiogenesis. Inhibition of RIP1 kinase activity is associated with caspase activation in infiltrating macrophages and decreased expression of proangiogenic M2-like markers but not M1-like markers. Similarly, in vitro, catalytic inhibition of RIP1 down-regulates the expression of M2-like markers in interleukin-4-activated bone marrow-derived macrophages, and this effect is blocked by simultaneous caspase inhibition. Collectively, these results demonstrate a nonnecrotic function of RIP1 kinase activity and suggest that RIP1-mediated modulation of macrophage activation may be a therapeutic target of pathological angiogenesis.


Subject(s)
GTPase-Activating Proteins/physiology , Macrophages/physiology , Neovascularization, Pathologic/enzymology , Animals , Biomarkers , Caspases/metabolism , Cells, Cultured , Collagen , Corneal Injuries/chemically induced , Corneal Injuries/etiology , Corneal Neovascularization/enzymology , Corneal Neovascularization/etiology , Corneal Neovascularization/pathology , Corneal Neovascularization/prevention & control , Drug Combinations , Enzyme Activation , Fibroblast Growth Factor 2/pharmacology , GTPase-Activating Proteins/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , In Situ Nick-End Labeling , Indoles/pharmacology , Indoles/therapeutic use , Laminin , Lasers/adverse effects , Macrophages/classification , Mice , Mice, Inbred C57BL , Models, Animal , Neovascularization, Pathologic/pathology , Oligopeptides/pharmacology , Proteoglycans , RNA, Messenger/biosynthesis , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
4.
Am J Pathol ; 190(10): 2056-2066, 2020 10.
Article in English | MEDLINE | ID: mdl-32693061

ABSTRACT

Glaucoma is a frequent and devastating long-term complication following ocular trauma, including corneal surgery, open globe injury, chemical burn, and infection. Postevent inflammation and neuroglial remodeling play a key role in subsequent ganglion cell apoptosis and glaucoma. To this end, this study was designed to investigate the amplifying role of monocyte infiltration into the retina. By using three different ocular injury mouse models (corneal suture, penetrating keratoplasty, and globe injury) and monocyte fate mapping techniques, we show that ocular trauma or surgery can cause robust infiltration of bone marrow-derived monocytes into the retina and subsequent neuroinflammation by up-regulation of Tnf, Il1b, and Il6 mRNA within 24 hours. This is accompanied by ganglion cell apoptosis and neurodegeneration. Prompt inhibition of tumor necrosis factor-α or IL-1ß markedly suppresses monocyte infiltration and ganglion cell loss. Thus, acute ocular injury (surgical or trauma) can lead to rapid neuroretinal inflammation and subsequent ganglion cell loss, the hallmark of glaucoma. Infiltrating monocytes play a central role in this process, likely amplifying the inflammatory cascade, aiding in the activation of retinal microglia. Prompt administration of cytokine inhibitors after ocular injury prevents this infiltration and ameliorates the damage to the retina-suggesting that it may be used prophylactically for neuroprotection against post-traumatic glaucoma.


Subject(s)
Cytokines/antagonists & inhibitors , Glaucoma/metabolism , Monocytes/pathology , Neuroglia/pathology , Retina/surgery , Animals , Burns, Chemical/metabolism , Burns, Chemical/pathology , Cornea/metabolism , Cornea/pathology , Disease Models, Animal , Glaucoma/pathology , Mice, Transgenic , Monocytes/metabolism , Retina/metabolism
5.
J Immunol ; 202(2): 539-549, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30541880

ABSTRACT

Reactive microglia and infiltrating peripheral monocytes have been implicated in many neurodegenerative diseases of the retina and CNS. However, their specific contribution in retinal degeneration remains unclear. We recently showed that peripheral monocytes that infiltrate the retina after ocular injury in mice become permanently engrafted into the tissue, establishing a proinflammatory phenotype that promotes neurodegeneration. In this study, we show that microglia regulate the process of neuroglia remodeling during ocular injury, and their depletion results in marked upregulation of inflammatory markers, such as Il17f, Tnfsf11, Ccl4, Il1a, Ccr2, Il4, Il5, and Csf2 in the retina, and abnormal engraftment of peripheral CCR2+ CX3CR1+ monocytes into the retina, which is associated with increased retinal ganglion cell loss, retinal nerve fiber layer thinning, and pigmentation onto the retinal surface. Furthermore, we show that other types of ocular injuries, such as penetrating corneal trauma and ocular hypertension also cause similar changes. However, optic nerve crush injury-mediated retinal ganglion cell loss evokes neither peripheral monocyte response in the retina nor pigmentation, although peripheral CX3CR1+ and CCR2+ monocytes infiltrate the optic nerve injury site and remain present for months. Our study suggests that microglia are key regulators of peripheral monocyte infiltration and retinal pigment epithelium migration, and their depletion results in abnormal neuroglia remodeling that exacerbates neuroretinal tissue damage. This mechanism of retinal damage through neuroglia remodeling may be clinically important for the treatment of patients with ocular injuries, including surgical traumas.


Subject(s)
Cornea/physiology , Eye Injuries/immunology , Microglia/physiology , Monocytes/physiology , Neurodegenerative Diseases/immunology , Neuroglia/physiology , Optic Nerve Injuries/immunology , Retina/physiology , Retinal Degeneration/immunology , Animals , Cell Movement , Cornea/pathology , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Animal , Neuronal Plasticity , Retina/pathology
6.
Proc Natl Acad Sci U S A ; 115(48): E11359-E11368, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30442669

ABSTRACT

Previous studies have demonstrated that ocular injury can lead to prompt infiltration of bone-marrow-derived peripheral monocytes into the retina. However, the ability of these cells to integrate into the tissue and become microglia has not been investigated. Here we show that such peripheral monocytes that infiltrate into the retina after ocular injury engraft permanently, migrate to the three distinct microglia strata, and adopt a microglia-like morphology. In the absence of ocular injury, peripheral monocytes that repopulate the retina after depletion with colony-stimulating factor 1 receptor (CSF1R) inhibitor remain sensitive to CSF1R inhibition and can be redepleted. Strikingly, consequent to ocular injury, the engrafted peripheral monocytes are resistant to depletion by CSF1R inhibitor and likely express low CSF1R. Moreover, these engrafted monocytes remain proinflammatory, expressing high levels of MHC-II, IL-1ß, and TNF-α over the long term. The observed permanent neuroglia remodeling after injury constitutes a major immunological change that may contribute to progressive retinal degeneration. These findings may also be relevant to other degenerative conditions of the retina and the central nervous system.


Subject(s)
Eye Injuries/immunology , Monocytes/immunology , Neuroglia/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Retina/immunology , Small Molecule Libraries/pharmacology , Animals , Cell Differentiation/drug effects , Eye Injuries/genetics , Eye Injuries/physiopathology , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Mice , Monocytes/cytology , Monocytes/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Retina/drug effects
7.
Am J Pathol ; 188(7): 1580-1596, 2018 07.
Article in English | MEDLINE | ID: mdl-29630857

ABSTRACT

Eyes that have experienced alkali burn to the surface are excessively susceptible to subsequent severe glaucoma and retinal ganglion cell loss, despite maximal efforts to prevent or slow down the disease. Recently, we have shown, in mice and rabbits, that such retinal damage is neither mediated by the alkali itself reaching the retina nor by intraocular pressure elevation. Rather, it is caused by the up-regulation of tumor necrosis factor-α (TNF-α), which rapidly diffuses posteriorly, causing retinal ganglion cell apoptosis and CD45+ cell activation. Herein, we investigated the involvement of peripheral blood monocytes and microglia in retinal damage. Using CX3CR1+/EGFP::CCR2+/RFP reporter mice and bone marrow chimeras, we show that peripheral CX3CR1+CD45hiCD11b+MHC-II+ monocytes infiltrate into the retina from the optic nerve at 24 hours after the burn and release further TNF-α. A secondary source of peripheral monocyte response originates from a rare population of patrolling myeloid CCR2+ cells of the retina that differentiate into CX3CR1+ macrophages within hours after the injury. As a result, CX3CR1+CD45loCD11b+ microglia become reactive at 7 days, causing further TNF-α release. Prompt TNF-α inhibition after corneal burn suppresses monocyte infiltration and microglia activation, and protects the retina. This study may prove relevant to other injuries of the central nervous system.


Subject(s)
Burns, Chemical/complications , Corneal Injuries/chemically induced , Corneal Injuries/complications , Disease Models, Animal , Microglia/pathology , Monocytes/pathology , Retinal Diseases/pathology , Animals , Mice , Microglia/immunology , Microglia/metabolism , Monocytes/immunology , Monocytes/metabolism , Rabbits , Retinal Diseases/etiology , Retinal Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Exp Eye Res ; 181: 136-144, 2019 04.
Article in English | MEDLINE | ID: mdl-30707890

ABSTRACT

N-methyl-D-aspartate (NMDA)-induced excitotoxicity is an acute form of experimental retinal injury as a result of overactivation of glutamate receptors. NLRP3 (nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain containing-3) inflammasome, one of the most studied sensors of innate immunity, has been reported to play a critical role in retinal neurodegeneration with controversial implications regarding neuroprotection and cell death. Thus far, it has not been elucidated whether NMDA-mediated excitotoxicity can trigger NLRP3 inflammasome in vivo. Moreover, it is unknown if NLRP3 is beneficial or detrimental to NMDA-mediated retinal cell death. Here, we employed a murine model of NMDA-induced retinal excitotoxicity by administering 100 nmoles of NMDA intravitreally, which resulted in massive TUNEL+ (TdT-dUTP terminal nick-end labelling) cell death in all retinal layers and especially in retinal ganglion cells (RGCs) 24 h post injection. NMDA insult in the retina potentiates macrophage/microglia cell infiltration, primes the NLRP3 inflammasome in a transcription-dependent manner and induces the expression of interleukin-1ß (IL-1ß). However, despite NLRP3 inflammasome upregulation, systemic deletion of Nlrp3 or Casp1 (caspase-1) did not significantly alter the NMDA-induced, excitotoxicity-mediated TUNEL+ retinal cell death at 24 h (acute phase). Similarly, the deletion of the two aforementioned genes did not alter the survival of the Brn3a+ (brain-specific homeobox/POU domain protein 3A) RGCs in a significant way at 3- or 7-days post injection (long-term phase). Our results indicate that NMDA-mediated retinal excitotoxicity induces immune cell recruitment and NLRP3 inflammasome activity even though inflammasome-mediated neuroinflammation is not a leading contributing factor to cell death in this type of retinal injury.


Subject(s)
Inflammasomes/metabolism , N-Methylaspartate/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Retinal Diseases , Animals , Cell Death , Disease Models, Animal , Interleukin-1beta/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Microglia/pathology , Retinal Diseases/chemically induced , Retinal Diseases/immunology , Retinal Diseases/metabolism
10.
Am J Pathol ; 187(6): 1327-1342, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28412300

ABSTRACT

Alkali burns to the eye constitute a leading cause of worldwide blindness. In recent case series, corneal transplantation revealed unexpected damage to the retina and optic nerve in chemically burned eyes. We investigated the physical, biochemical, and immunological components of retinal injury after alkali burn and explored a novel neuroprotective regimen suitable for prompt administration in emergency departments. Thus, in vivo pH, oxygen, and oxidation reduction measurements were performed in the anterior and posterior segment of mouse and rabbit eyes using implantable microsensors. Tissue inflammation was assessed by immunohistochemistry and flow cytometry. The experiments confirmed that the retinal damage is not mediated by direct effect of the alkali, which is effectively buffered by the anterior segment. Rather, pH, oxygen, and oxidation reduction changes were restricted to the cornea and the anterior chamber, where they caused profound uveal inflammation and release of proinflammatory cytokines. The latter rapidly diffuse to the posterior segment, triggering retinal damage. Tumor necrosis factor-α was identified as a key proinflammatory mediator of retinal ganglion cell death. Blockade, by either monoclonal antibody or tumor necrosis factor receptor gene knockout, reduced inflammation and retinal ganglion cell loss. Intraocular pressure elevation was not observed in experimental alkali burns. These findings illuminate the mechanism by which alkali burns cause retinal damage and may have importance in designing therapies for retinal protection.


Subject(s)
Burns, Chemical/metabolism , Eye Burns/metabolism , Retina/injuries , Alkalies , Animals , Apoptosis/drug effects , Apoptosis/physiology , Burns, Chemical/drug therapy , Burns, Chemical/etiology , Burns, Chemical/pathology , Cornea/immunology , Corneal Injuries/drug therapy , Corneal Injuries/etiology , Corneal Injuries/metabolism , Corneal Injuries/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Eye Burns/drug therapy , Eye Burns/etiology , Eye Burns/pathology , Hydrogen-Ion Concentration , Infliximab/pharmacology , Infliximab/therapeutic use , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidation-Reduction , Rabbits , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Retina/immunology , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Sodium Hydroxide , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Uvea/metabolism , Uveitis, Anterior/chemically induced , Uveitis, Anterior/metabolism , Uveitis, Anterior/pathology , Uveitis, Anterior/prevention & control
11.
Transl Vis Sci Technol ; 13(5): 2, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38696181

ABSTRACT

Purpose: Currently, no solution exists to restore natural eyelid kinematics for patients with complete eyelid paralysis due to loss of function of both the levator palpebrae superioris and orbicularis oculi. These rare cases are prone to complications of chronic exposure keratopathy which may lead to corneal blindness. We hypothesized that magnetic force could be used to fully automate eyelid movement in these cases through the use of eyelid-attached magnets and a spectacle-mounted magnet driven by a programmable motor (motorized magnetic levator prosthesis [MMLP]). Methods: To test this hypothesis and establish proof of concept, we performed a finite element analysis (FEA) for a prototype MMLP to check the eyelid-opening force generated by the device and verified the results with experimental measurements in a volunteer with total bidirectional eyelid paralysis. The subject was then fitted with a prototype to check the performance of the device and its success. Results: With MMLP, eye opening was restored to near normal, and blinking was fully automated in close synchrony with the motor-driven polarity reversal, with full closure on the blink. The device was well tolerated, and the participant was pleased with the comfort and performance. Conclusions: FEA simulation results conformed to the experimentally observed trend, further supporting the proof of concept and design parameters. This is the first viable approach in human patients with proof of concept for complete reanimation of a bidirectionally paretic eyelid. Further study is warranted to refine the prototype and determine the feasibility and safety of prolonged use. Translational Relevance: This is first proof of concept for our device for total bidirectional eyelid paralysis.


Subject(s)
Blinking , Eyelids , Proof of Concept Study , Humans , Blinking/physiology , Eyelids/physiopathology , Eyelid Diseases/physiopathology , Eyelid Diseases/therapy , Oculomotor Muscles/physiopathology , Finite Element Analysis , Biomechanical Phenomena , Prostheses and Implants , Prosthesis Design , Magnets , Male
12.
Acta Ophthalmol ; 102(3): e381-e394, 2024 May.
Article in English | MEDLINE | ID: mdl-37803488

ABSTRACT

BACKGROUND AND PURPOSE: Late secondary glaucoma is an often-severe complication after acute events like anterior segment surgery, trauma and infection. TNF-α is a major mediator that is rapidly upregulated, diffusing also to the retina and causes apoptosis of the ganglion cells and degeneration of their optic nerve axons (mediating steps to glaucomatous damage). Anti-TNF-α antibodies are in animals very effective in protecting the retinal cells and the optic nerve-and might therefore be useful prophylactically against secondary glaucoma in future such patients. Here we evaluate (1) toxicity and (2) efficacy of two TNF-α inhibitors (adalimumab and infliximab), in rabbits by subconjunctival administration. METHODS: For drug toxicity, animals with normal, unburned corneas were injected with adalimumab (0.4, 4, or 40 mg), or infliximab (1, 10, or 100 mg). For drug efficacy, other animals were subjected to alkali burn before such injection, or steroids (for control). The rabbits were evaluated clinically with slit lamp and photography, electroretinography, optical coherence tomography, and intraocular pressure manometry. A sub-set of eyes were stained ex vivo after 3 days for retinal cell apoptosis (TUNEL). In other experiments the optic nerves were evaluated by paraphenylenediamine staining after 50 or 90 days. Loss of retinal cells and optic nerve degeneration were quantified. RESULTS: Subconjunctival administration of 0.4 mg or 4.0 mg adalimumab were well tolerated, whereas 40.0 mg was toxic to the retina. 1, 10, or 100 mg infliximab were also well tolerated. Analysis of the optic nerve axons after 50 days confirmed the safety of 4.0 mg adalimumab and of 100 mg infliximab. For efficacy, 4.0 mg adalimumab subconjunctivally in 0.08 mL provided practically full protection against retinal cell apoptosis 3 days following alkali burn, and infliximab 100 mg only slightly less. At 90 days following burn injury, control optic nerves showed about 50% axon loss as compared to 8% in the adalimumab treatment group. CONCLUSIONS: Subconjunctival injection of 4.0 mg adalimumab in rabbits shows no eye toxicity and provides excellent neuroprotection, both short (3 days) and long-term (90 days). Our total. accumulated data from several of our studies, combined with the present paper, suggest that corneal injuries, including surgery, might benefit from routine administration of anti-TNF-α biologics to reduce inflammation and future secondary glaucoma.


Subject(s)
Axons , Burns, Chemical , Cornea , Optic Nerve , Tumor Necrosis Factor Inhibitors , Animals , Rabbits , Adalimumab/therapeutic use , Apoptosis , Burns, Chemical/drug therapy , Disease Models, Animal , Glaucoma , Infliximab/therapeutic use , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha
13.
Transl Vis Sci Technol ; 12(8): 11, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37566397

ABSTRACT

Purpose: Blepharoptosis is a common oculoplastic condition causing incomplete opening of the upper eyelid. Surgical approaches, the mainstay for correction, often fail to improve blink function. The purpose of this study was to develop a nonsurgical treatment option for severe ptosis that allows blink re-animation. Methods: Magnetic force required to perform blink re-animation was characterized by evaluation of eye-opening and closing using inter-palpebral fissure (IPF) outcomes with various combinations of eyelid array and box magnets. Optimal size of the spectacle magnet that achieved forces required for optimal blink dynamics was selected using simulation. The adjustable magnetic levator prosthesis (aMLP) included an eyelid array magnet and an adjustable rotating spectacle magnet that allowed change in the magnetic direction, thus changing the net magnetic interactive force between the magnets. The clinical feasibility of aMLP in improving eye opening without limiting eye closing was evaluated in patients with ptosis through a proof-of-concept study using IPF and comfort outcomes. Results: Optimal eye opening and closing was achieved by a magnet-array combination providing 45 grams of surface force (gF) in the tested ptosis population. The aMLP was able to modulate eye opening and closing with change in rotation of the spectacle magnet in two patients with ptotis. The best fitting of an aMLP improved IPF opening without limiting eye closing and with good comfort reported. Conclusions: Preliminary results suggest that the an aMLP can correct ptosis without adversely affecting blink function. Further evaluation in a larger patient population is warranted. Translational Relevance: A nonsurgical, proof of concept, adjustable magnetic treatment option for blink re-animation in patients with severe ptosis is presented.


Subject(s)
Blepharoptosis , Humans , Blepharoptosis/surgery , Eyelids/surgery , Prostheses and Implants , Magnetic Phenomena
14.
Transl Vis Sci Technol ; 12(12): 1, 2023 12 01.
Article in English | MEDLINE | ID: mdl-38038607

ABSTRACT

Purpose: Examine the effect of force modulation via angular translation of a static magnetic field for customizable treatment of severe blepharoptosis. Methods: Prototype adjustable-force magnetic levator prostheses (aMLP) consisted of a spectacle-mounted magnet in rotatable housing and small eyelid-attached magnets embedded in a biocompatible polymer. Interpalpebral fissure (IPF) of 17 participants with severe blepharoptosis was continuously measured for one minute at five spectacle magnet angles, with order randomized and participant and data analyst masked. The hypothesis that angular position affected opening IPF (o-IPF), minimum blink IPF (m-IPF), and comfort ratings (1-10) was tested. Results: The aMLP improved o-IPF from 4.5 mm without the device to 6.2 mm on the lowest force setting (P < 0.001) and 7.1 mm on the highest setting (P < 0.001) and allowed for complete volitional blink regardless of setting (average m-IPF 0.4 mm and no change with aMLP; P = 0.76). Spontaneous blink without the device (2.0 mm) was affected on the highest force setting (m-IPF 3.9 mm; P < 0.001) but only marginally so on the lowest setting (3.0 mm; P = 0.06). Comfort (7.6/10) did not vary with the angle (P > 0.36). Profile analysis found substantial individual responses to angle (P < 0.001), confirming the value of customization. Conclusions: Angular translation provided adjustable force, which had a statistically and clinically meaningful impact on eye opening and the completeness of the spontaneous blink. This quantitative evidence supports continued use of the angular translation mechanism for force adjustment in the customizable magnetic correction of severe blepharoptosis. Translational Relevance: Evidence for the benefit of customizable magnetic force via angular translation in a larger sample of participants than reported previously.


Subject(s)
Blepharoptosis , Humans , Blepharoptosis/surgery , Eyelids , Prostheses and Implants , Magnetic Phenomena
15.
Res Sq ; 2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37720036

ABSTRACT

Colony-stimulating factor 1 receptor (CSF1R) inhibition has been proposed as a specific method for microglia depletion. However, recent work revealed that in addition to microglia, CSF1R inhibition also affects other innate immune cells, such as peripheral monocytes and tissue-resident macrophages of the lung, liver, spleen, and peritoneum. Here, we show that this effect is not restricted to innate immune cells only but extends to the adaptive immune compartment. CSF1R inhibition alters the transcriptional profile of bone marrow cells that control T helper cell activation. In vivo or ex vivo inhibition of CSF1R profoundly changes the transcriptional profile of CD4+ cells and suppresses Th1 and Th2 differentiation in directionally stimulated and unstimulated cells and independently of microglia depletion. Given that T cells also contribute in CNS pathology, these effects may have practical implications in the interpretation of relevant experimental data.

16.
Pharmaceutics ; 15(8)2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37631272

ABSTRACT

PURPOSE: This study aimed to develop a clinically feasible and practical therapy for multi-ocular protection following ocular injury by using a thermosensitive drug delivery system (DDS) for sustained delivery of TNF-α and VEGF inhibitors to the eye. METHODS: A thermosensitive, biodegradable hydrogel DDS (PLGA-PEG-PLGA triblock polymer) loaded with 0.7 mg of adalimumab and 1.4 mg of aflibercept was injected subconjunctivally into Dutch-belted pigmented rabbits after corneal alkali injury. Control rabbits received 2 mg of IgG-loaded DDS or 1.4 mg of aflibercept-loaded DDS. Animals were followed for 3 months and assessed for tolerability and prevention of corneal neovascularization (NV), improvement of corneal re-epithelialization, inhibition of retinal ganglion cell (RGC) and optic nerve axon loss, and inhibition of immune cell infiltration into the cornea. Drug-release kinetics was assessed in vivo using an aqueous humor protein analysis. RESULTS: A single subconjunctival administration of dual anti-TNF-α/anti-VEGF DDS achieved a sustained 3-month delivery of antibodies to the anterior chamber, iris, ciliary body, and retina. Administration after corneal alkali burn suppressed CD45+ immune cell infiltration into the cornea, completely inhibited cornea NV for 3 months, accelerated corneal re-epithelialization and wound healing, and prevented RGC and optic nerve axon loss at 3 months. In contrast, anti-VEGF alone or IgG DDS treatment led to persistent corneal epithelial defect (combined: <1%; anti-VEGF: 15%; IgG: 10%, of cornea area), increased infiltration of CD45+ immune cells into the cornea (combined: 28 ± 20; anti-VEGF: 730 ± 178; anti-IgG: 360 ± 186, cells/section), and significant loss of RGCs (combined: 2.7%; anti-VEGF: 63%; IgG: 45%) and optic nerve axons at 3 months. The aqueous humor protein analysis showed first-order release kinetics without adverse effects at the injection site. CONCLUSIONS: Concomitant inhibition of TNF-α and VEGF prevents corneal neovascularization and ameliorates subsequent irreversible damage to the retina and optic nerve after severe ocular injury. A single subconjunctival administration of this therapy, using a biodegradable, slow-release thermosensitive DDS, achieved the sustained elution of therapeutic levels of antibodies to all ocular tissues for 3 months. This therapeutic approach has the potential to dramatically improve the outcomes of severe ocular injuries in patients and improve the therapeutic outcomes in patients with retinal vascular diseases.

17.
Cells ; 12(16)2023 08 18.
Article in English | MEDLINE | ID: mdl-37626899

ABSTRACT

Limbal stem cell (LSC) deficiency is a frequent and severe complication after chemical injury to the eye. Previous studies have assumed this is mediated directly by the caustic agent. Here we show that LSC damage occurs through immune cell mediators, even without direct injury to LSCs. In particular, pH elevation in the anterior chamber (AC) causes acute uveal stress, the release of inflammatory cytokines at the basal limbal tissue, and subsequent LSC damage and death. Peripheral C-C chemokine receptor type 2 positive/CX3C motif chemokine receptor 1 negative (CCR2+ CX3CR1-) monocytes are the key mediators of LSC damage through the upregulation of tumor necrosis factor-alpha (TNF-α) at the limbus. In contrast to peripherally derived monocytes, CX3CR1+ CCR2- tissue-resident macrophages have a protective role, and their depletion prior to injury exacerbates LSC loss and increases LSC vulnerability to TNF-α-mediated apoptosis independently of CCR2+ cell infiltration into the tissue. Consistently, repopulation of the tissue by new resident macrophages not only restores the protective M2-like phenotype of macrophages but also suppresses LSC loss after exposure to inflammatory signals. These findings may have clinical implications in patients with LSC loss after chemical burns or due to other inflammatory conditions.


Subject(s)
Eye Injuries , Limbal Stem Cell Deficiency , Humans , Monocytes , Limbal Stem Cells , Tumor Necrosis Factor-alpha , Macrophages , Receptors, Chemokine
18.
Eye (Lond) ; 37(10): 2117-2125, 2023 07.
Article in English | MEDLINE | ID: mdl-36329167

ABSTRACT

OBJECTIVE: To assess the cumulative incidence and risk factors for glaucoma development and progression within 1-2 years following corneal transplant surgery. DESIGN: Retrospective cohort study. METHODS: Patients undergoing penetrating keratoplasty (PK), deep anterior lamellar keratoplasty (DALK), Descemet stripping endothelial keratoplasty (DSEK), Descemet membrane endothelial keratoplasty (DMEK), Boston keratoprosthesis type I (KPro) implantation, or endothelial keratoplasty (DSEK or DMEK) under previous PK (EK under previous PK) at one academic institution with at least 1 year of follow-up were included. Primary outcome measures were cumulative incidence of glaucoma development and progression after corneal transplant, in patients without and with preoperative glaucoma, respectively. Risk factors for glaucoma development and progression were also assessed. RESULTS: Four hundred and thirty-one eyes of 431 patients undergoing PK (113), DALK (17), DSEK (71), DMEK (168), KPro (35) and EK under previous PK (27) with a mean follow-up of 22.9 months were analyzed. The 1-year cumulative incidence for glaucoma development and progression was 28.0% and 17.8% in patients without and with preoperative glaucoma, respectively. In a Cox proportional hazards analysis, DSEK surgery, KPro implantation, average intraocular pressure (IOP) through follow-up and postoperative IOP spikes of ≥30 mmHg were each independently associated with glaucoma development or progression (p < 0.04 for all). CONCLUSIONS: A significant proportion of patients developed glaucoma or exhibited glaucoma progression within 1 year after corneal transplantation. Patient selection for DSEK may partly explain the higher risk for glaucoma in these patients. Postoperative IOP spikes should be minimized and may indicate the need for co-management with a glaucoma specialist.


Subject(s)
Corneal Diseases , Descemet Stripping Endothelial Keratoplasty , Glaucoma , Humans , Incidence , Retrospective Studies , Cornea , Corneal Diseases/epidemiology , Corneal Diseases/surgery , Corneal Diseases/complications , Descemet Stripping Endothelial Keratoplasty/adverse effects , Prostheses and Implants/adverse effects , Glaucoma/epidemiology , Glaucoma/etiology , Glaucoma/surgery , Keratoplasty, Penetrating/adverse effects , Risk Factors , Follow-Up Studies
19.
Aesthetic Plast Surg ; 36(3): 546-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22101985

ABSTRACT

BACKGROUND: The purpose of this study was to present an alternative surgical procedure for the correction of persistent malar bags and to review the literature. METHODS: A 45-year-old female patient with persistent malar bags, secondary to previous blepharoplasty, was admitted. Liposuction of the edema and suspension of the orbicularis muscle to the temporal region was performed through a lateral canthal incision. RESULTS: A successful malar bag removal was demonstrated, exhibiting stable results at the 6-month follow-up. CONCLUSION: Persistent malar bags resulting from previous blepharoplasty may be difficult to correct with conventional treatment. The combination of liposuction and suspension of the orbicularis oculi is proposed as an efficient alternative for the correction of persistent malar bags.


Subject(s)
Blepharoplasty/methods , Lipectomy , Female , Humans , Middle Aged , Oculomotor Muscles
20.
Transl Vis Sci Technol ; 11(10): 34, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36269183

ABSTRACT

Purpose: To determine the feasibility of a custom frame generation approach for nonsurgical management of severe blepharoptosis with the magnetic levator prosthesis (MLP). Methods: Participants (n = 8) with severe blepharoptosis (obscuring the visual axis) in one or both eyes who had previously been using a non-custom MLP had a craniofacial scan with a smartphone app to generate a custom MLP frame. A magnetic adhesive was attached to the affected eyelid. The custom MLP frame held a cylindrical magnet near the eyebrow above the affected eyelid, suspending it in the magnetic field while still allowing blinking. The spectacle magnet could be rotated manually, providing adjustable force via angular translation of the magnetic field. Fitting success and comfort were recorded, and interpalpebral fissure (IPF) was measured from video frames after 20 minutes in-office and one-week at-home use. Preference was documented, custom versus non-custom. Results: Overall, 88% of patients (7/8) were successfully fitted with a median 9/10 comfort (interquartile 7-10) and median ptosis improvement of 2.3 mm (1.3-5.0); P = 0.01). Exact binomial testing suggested, with 80% power, that the true population success rate was significantly greater than 45% (P = 0.05). Five participants took the custom MLP home for one week, with only one case of mild conjunctival redness which resolved without treatment. Highest to lowest force modulation resulted in a marginally significant median IPF adjustment of 1.5 mm (0.8 to 2.7; P = 0.06). All preferred the custom frame. Conclusions: The three-dimensional custom MLP frame generation approach using a smartphone app-based craniofacial scan is a feasible approach for clinical deployment of the MLP. Translational Relevance: First demonstration of customized frame generation for the MLP.


Subject(s)
Artificial Limbs , Blepharoptosis , Humans , Blepharoptosis/surgery , Oculomotor Muscles/diagnostic imaging , Oculomotor Muscles/surgery , Feasibility Studies , Printing, Three-Dimensional , Magnetic Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL