Network


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

Hotspot


Dive into the research topics where Kimberly Dine is active.

Publication


Featured researches published by Kimberly Dine.


Frontiers in Neurology | 2012

Resveratrol neuroprotection in a chronic mouse model of multiple sclerosis

Zoe Fonseca-Kelly; Mayssa Nassrallah; Jorge Uribe; Reas S. Khan; Kimberly Dine; Mahasweta Dutt; Kenneth S. Shindler

Resveratrol is a naturally occurring polyphenol that activates SIRT1, an NAD-dependent deacetylase. SRT501, a pharmaceutical formulation of resveratrol with enhanced systemic absorption, prevents neuronal loss without suppressing inflammation in mice with relapsing experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). In contrast, resveratrol has been reported to suppress inflammation in chronic EAE, although neuroprotective effects were not evaluated. The current studies examine potential neuroprotective and immunomodulatory effects of resveratrol in chronic EAE induced by immunization with myelin oligodendroglial glycoprotein peptide in C57/Bl6 mice. Effects of two distinct formulations of resveratrol administered daily orally were compared. Resveratrol delayed the onset of EAE compared to vehicle-treated EAE mice, but did not prevent or alter the phenotype of inflammation in spinal cords or optic nerves. Significant neuroprotective effects were observed, with higher numbers of retinal ganglion cells found in eyes of resveratrol-treated EAE mice with optic nerve inflammation. Results demonstrate that resveratrol prevents neuronal loss in this chronic demyelinating disease model, similar to its effects in relapsing EAE. Differences in immunosuppression compared with prior studies suggest that immunomodulatory effects may be limited and may depend on specific immunization parameters or timing of treatment. Importantly, neuroprotective effects can occur without immunosuppression, suggesting a potential additive benefit of resveratrol in combination with anti-inflammatory therapies for MS.


Investigative Ophthalmology & Visual Science | 2013

SIRT1 Promotes RGC Survival and Delays Loss of Function Following Optic Nerve Crush

Ling Zuo; Reas S. Khan; Vivian S. Lee; Kimberly Dine; Wen Wu; Kenneth S. Shindler

PURPOSE Activation of SIRT1 deacetylase prevents retinal ganglion cell (RGC) loss in experimental optic neuritis, an inflammatory optic neuropathy. While mechanisms of this effect are not known, evidence suggests it involves reduction of oxidative stress. We hypothesized that SIRT1 reduces RGC loss due to oxidative stress in noninflammatory optic neuropathies, and examined effects following traumatic injury. METHODS Optic nerve crush injury was induced in wild-type C57BL/6 mice, mice overexpressing SIRT1, and mice with conditional deletion of SIRT1 in neurons. Wild-type mice were treated daily with vehicle or 250 mg/kg resveratrol, a naturally occurring polyphenol that activates SIRT1. RGC function was assessed by pupillometry and optokinetic responses (OKR), and RGC survival was measured. Superoxide levels were measured to assess oxidative stress. RESULTS Significant decreases in pupillary light responses, OKR and RGC survival occurred 1 week after optic nerve crush, with progressive worsening at 2 to 4 weeks. Resveratrol treatment and SIRT1 overexpression delayed RGC loss and loss of pupillary light responses following optic nerve crush, although no change in RGC loss occurred in neuronal SIRT1-deficient mice. A significant accumulation of superoxide was detected in wild-type optic nerves following crush, and was reduced in mice overexpressing SIRT1 or treated with resveratrol. CONCLUSIONS SIRT1 delays RGC loss following traumatic injury. Effects are associated with reduced oxidative stress. Results suggest SIRT1-activating drugs may have a specific role in preventing traumatic optic nerve damage, and suggest a broader role for this strategy in treating a variety of optic neuropathies that may include a component of oxidative stress.


Investigative Ophthalmology & Visual Science | 2014

HE3286 reduces axonal loss and preserves retinal ganglion cell function in experimental optic neuritis.

Reas S. Khan; Kimberly Dine; Esteban Luna; Clarence Ahlem; Kenneth S. Shindler

PURPOSE Optic nerve inflammation, demyelination, and axonal loss are all prominent features of optic neuritis. While corticosteroids hasten visual recovery in optic neuritis, no treatment improves final visual outcomes. HE3286 (17α-ethynyl-5-androstene-3β,7β,17β-triol), a synthetic derivative of a natural steroid, β-AET (5-androstene-3β,7β,17β-triol), exerts anti-inflammatory effects in several disease models and has purported neuroprotective effects as well. HE3286s ability to suppress optic neuritis was examined in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. METHODS Experimental autoimmune encephalomyelitis was induced in C57/BL6 mice. Mice were treated daily with intraperitoneal vehicle or 40 mg/kg HE3286. Visual function was assessed by optokinetic responses (OKR) at baseline and every 10 days until euthanasia at 40 days post immunization. Retinas and optic nerves were isolated. Inflammation (hematoxylin and eosin and Iba1 staining), demyelination (Luxol fast blue staining), and axonal loss (neurofilament staining) were assessed in optic nerve sections. Retinal ganglion cells (RGCs) were immunolabeled with Brn3a antibodies to quantify RGC survival. RESULTS Progressive decreases in OKR occurred in vehicle-treated EAE mice, and HE3286 treatment reduced the level of this vision loss. HE3286 also attenuated the degree of inflammation, demyelination, and axonal loss in EAE optic nerves as compared to nerves from vehicle-treated EAE mice. Retinal ganglion cell loss that occurred in both vehicle- and HE3286-treated EAE mice was reduced in the temporal retinal quadrant of HE3286-treated mice. CONCLUSIONS HE3286 suppresses inflammation, reduces demyelination and axonal loss, and promotes RGC survival during experimental optic neuritis. Importantly, HE3286 treatment also preserves some RGC function. Results suggest that HE3286 is a potential novel treatment for optic neuritis.


Scientific Reports | 2017

Intranasal Delivery of A Novel Amnion Cell Secretome Prevents Neuronal Damage and Preserves Function In A Mouse Multiple Sclerosis Model

Reas S. Khan; Kimberly Dine; Bailey Bauman; Michael Lorentsen; Lisa Lin; Helayna Brown; Leah R. Hanson; Aleta L. Svitak; Howard Wessel; Larry R. Brown; Kenneth S. Shindler

The ability of a novel intranasally delivered amnion cell derived biologic to suppress inflammation, prevent neuronal damage and preserve neurologic function in the experimental autoimmune encephalomyelitis animal model of multiple sclerosis was assessed. Currently, there are no existing optic nerve treatment methods for disease or trauma that result in permanent vision loss. Demyelinating optic nerve inflammation, termed optic neuritis, induces permanent visual dysfunction due to retinal ganglion cell damage in multiple sclerosis and experimental autoimmune encephalomyelitis. ST266, the biological secretome of Amnion-derived Multipotent Progenitor cells, contains multiple anti-inflammatory cytokines and growth factors. Intranasally administered ST266 accumulated in rodent eyes and optic nerves, attenuated visual dysfunction, and prevented retinal ganglion cell loss in experimental optic neuritis, with reduced inflammation and demyelination. Additionally, ST266 reduced retinal ganglion cell death in vitro. Neuroprotective effects involved oxidative stress reduction, SIRT1-mediated mitochondrial function promotion, and pAKT signaling. Intranasal delivery of neuroprotective ST266 is a potential novel, noninvasive therapeutic modality for the eyes, optic nerves and brain. The unique combination of biologic molecules in ST266 provides an innovative approach with broad implications for suppressing inflammation in autoimmune diseases, and for preventing neuronal damage in acute neuronal injury and chronic neurodegenerative diseases such as multiple sclerosis.


Oxidative Medicine and Cellular Longevity | 2017

Mitochondrial Uncoupler Prodrug of 2,4-Dinitrophenol, MP201, Prevents Neuronal Damage and Preserves Vision in Experimental Optic Neuritis

Reas S. Khan; Kimberly Dine; John G. Geisler; Kenneth S. Shindler

The ability of novel mitochondrial uncoupler prodrug of 2,4-dinitrophenol (DNP), MP201, to prevent neuronal damage and preserve visual function in an experimental autoimmune encephalomyelitis (EAE) model of optic neuritis was evaluated. Optic nerve inflammation, demyelination, and axonal loss are prominent features of optic neuritis, an inflammatory optic neuropathy often associated with the central nervous system demyelinating disease multiple sclerosis. Currently, optic neuritis is frequently treated with high-dose corticosteroids, but treatment fails to prevent permanent neuronal damage and associated vision changes that occur as optic neuritis resolves, thus suggesting that additional therapies are required. MP201 administered orally, once per day, attenuated visual dysfunction, preserved retinal ganglion cells (RGCs), and reduced RGC axonal loss and demyelination in the optic nerves of EAE mice, with limited effects on inflammation. The prominent mild mitochondrial uncoupling properties of MP201, with slow elimination of DNP, may contribute to the neuroprotective effect by modulating the entire mitochondrias physiology directly. Results suggest that MP201 is a potential novel treatment for optic neuritis.


Ophthalmic Plastic and Reconstructive Surgery | 2016

Histologic Evidence of Orbital Inflammation from Retrobulbar Alcohol and Chlorpromazine Injection: A Clinicopathologic Study in Human & Rat Orbits.

Kian Eftekhari; Kenneth S. Shindler; Vivian S. Lee; Kimberly Dine; Lauren A. Eckstein; M. Reza Vagefi

Purpose: Retrobulbar injections of alcohol and chlorpromazine are used for the treatment of blind, painful eyes. There have been reports of inflammation after retrobulbar injections of these agents, but the histologic effects are not well characterized. A clinical case with histopathologic confirmation of inflammation after retrobulbar alcohol injection led the authors to develop a rat model to examine these effects. Methods: Adult Lewis rats were given retrobulbar injections of either 0.1 ml of absolute alcohol or 25 mg/ml chlorpromazine in the right orbit, and 0.1 ml of saline in the left orbit as a control. Rats were euthanized, perfused, and postfixed at 1 to 2 weeks after injection. Exenterated orbital tissue was sectioned for histologic staining. Slides were reviewed by a masked ocular pathologist who evaluated the level of orbital inflammation. Results: Histopathology demonstrated foci of granulomatous inflammation in the orbit of the patient and similar inflammation in the rat orbits injected with retrobulbar alcohol. In the chlorpromazine group, only 1 rat demonstrated small foci of inflammation, while the control orbits injected with saline showed no inflammation. On blinded qualitative analysis, the orbits receiving retrobulbar alcohol had greater inflammation than the orbits receiving either saline or chlorpromazine. Conclusions: Our findings in this preclinical pilot study suggest that retrobulbar alcohol injections incite significant orbital inflammation, whereas retrobulbar chlorpromazine induces little or no inflammation. This potential inflammatory response should be considered when selecting an agent for pain management, particularly if future orbital surgery is anticipated.


Immunology Innovation | 2013

Decreased signal transducers and activators of transcription (STAT) protein expression in lymphatic organs during EAE development in mice.

Wen Xuan Wu; Ling Zuo; Kimberly Dine; Kenneth S. Shindler

Experimental autoimmune encephalomyelitis (EAE) is mediated by myelin-specific CD4+ T cells secreting Th1 and/or Th17 cytokines. Signal transducer and activator of transcription (STAT) family proteins have essential roles in transmitting Th1 and/or Th17 cytokine-mediated signals. However, most studies demonstrating the importance of the STAT signaling system in EAE have focused on distinct members of this family, often looking at their role specifically in the central nervous system, or in vitro. There is limited information available regarding the temporal and spatial expression patterns of each STAT protein and interplay between STAT proteins over the course of EAE development in critical lymphatic organs in vivo. In the present study, we demonstrate dramatic and progressive decrease of all six STAT family members (STAT1, STAT2, STAT3, STAT4, STAT5, STAT6) in the spleen and lymph nodes through the course of EAE development in SJL/J mice, in contrast with almost steady expression of thymic STAT proteins. Decreased splenic and lymphatic STAT expression was accompanied by significant enlargement of the spleen and lymph nodes, and histological proliferation of T cell areas with remodeling of the splenic microstructure in EAE mice. All STAT family members except STAT2 were mainly confined in T cell areas in spleen, whereas they were distributed in a protein specific manner in thymus. We present here a comprehensive analysis of all six members of the STAT family in spleen, lymph nodes and thymus through the development phase of EAE. Results suggest that EAE induced inflammatory T cells may develop distinct biological features different from normal splenic T cells due to altered STAT signaling.


Investigative Ophthalmology & Visual Science | 2018

SIRT1 and NRF2 Gene Transfer Mediate Distinct Neuroprotective Effects Upon Retinal Ganglion Cell Survival and Function in Experimental Optic Neuritis

Devin S. McDougald; Kimberly Dine; Alexandra Zezulin; Jean Bennett; Kenneth S. Shindler

Purpose Optic neuritis is a condition defined by autoimmune-mediated demyelination of the optic nerve and death of retinal ganglion cells. SIRT1 and NRF2 stimulate anti-inflammatory mechanisms and have previously demonstrated therapeutic value in preclinical models of neurodegenerative disease. Here we investigated the neuroprotective potential of SIRT1 or NRF2 gene transfer using adeno-associated virus (AAV) vectors in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Methods C57Bl/6J mice were administered intravitreal doses of AAV2 vectors and immunized to induce EAE symptoms. Visual function was examined by recording the optokinetic response (OKR) just prior to EAE induction and once every 7 days postinduction for 7 weeks. Retina and optic nerves were harvested to investigate retinal ganglion cell survival (immunolabeling with Brn3a antibodies); inflammation (hematoxylin and eosin staining); and demyelination (luxol fast blue staining). Results Animals modeling EAE demonstrate reduced visual acuity compared to sham-induced controls. Intravitreal delivery of AAV2-NRF2 did not preserve visual function. However, AAV2-SIRT1 mediated significant preservation of the OKR compared to AAV2-eGFP controls. Treatment with AAV2-NRF2 promoted RGC survival while AAV2-SIRT1 mediated an upward trend in protection compared to vehicle and AAV2-eGFP controls. Neither NRF2 nor SIRT1 gene augmentation was able to suppress optic nerve inflammation or demyelination. Conclusions AAV-mediated overexpression of NRF2 or SIRT1 within RGCs mediates distinct neuroprotective effects upon visual function and RGC survival. This study expands our understanding of SIRT1 and NRF2-mediated neuroprotection in the context of MS pathogenesis and optic neuropathies.


Investigative Ophthalmology & Visual Science | 2018

RGC Neuroprotection Following Optic Nerve Trauma Mediated By Intranasal Delivery of Amnion Cell Secretome

Gabriela A. Grinblat; Reas S. Khan; Kimberly Dine; Howard Wessel; Larry R. Brown; Kenneth S. Shindler

Purpose Intranasally delivered ST266, the biological, proteinaceous secretome of amnion-derived multipotent progenitor cells, reduces retinal ganglion cell (RGC) loss, optic nerve inflammation, and demyelination in experimental optic neuritis. This unique therapy and novel administration route delivers numerous cytokines and growth factors to the eye and optic nerve, suggesting a potential to also treat other optic neuropathies. Thus, ST266-mediated neuroprotection was examined following traumatic optic nerve injury. Methods Optic nerve crush injury was surgically induced in C57BL/6J mice. Mice were treated daily with intranasal PBS or ST266. RGC function was assessed by optokinetic responses (OKRs), RGCs were counted, and optic nerve sections were stained with luxol fast blue and anti-neurofilament antibodies to assess myelin and RGC axon damage. Results Intranasal ST266 administered daily for 5 days, beginning at the time that a 1-second optic nerve crush was performed, significantly attenuated OKR decreases. Furthermore, ST266 treatment reduced damage to RGC axons and myelin within optic nerves, and blocked RGC loss. Following a 4-second optic nerve crush, intranasal ST266 increased RGC survival and showed a trend toward reduced RGC axon and myelin damage. Ten days following optic nerve crush, ST266 prevented myelin damage, while also inducing a trend toward increased RGC survival and visual function. Conclusions ST266 significantly attenuates traumatic optic neuropathy. Neuroprotective effects of this unique combination of biologic molecules observed here and previously in optic neuritis suggest potential broad application for preventing neuronal damage in multiple optic nerve disorders. Furthermore, results support intranasal delivery as a novel, noninvasive therapeutic modality for eyes and optic nerves.


Frontiers in Cellular and Infection Microbiology | 2018

Intracranial Inoculation Is More Potent Than Intranasal Inoculation for Inducing Optic Neuritis in the Mouse Hepatitis Virus-Induced Model of Multiple Sclerosis

Manmeet Singh; Reas S. Khan; Kimberly Dine; Jayasri Das Sarma; Kenneth S. Shindler

Neurotropic strains of mouse hepatitis virus (MHV) induce acute inflammation and chronic demyelination in the spinal cord and optic nerves mediated by axonal spread following intracranial inoculation in mice, with pathologic features similar to the human demyelinating disease multiple sclerosis. Spinal cord demyelination is also induced following intranasal inoculation with neurotropic MHV strains, however much higher viral doses are required as compared to intracranial inoculation. Recently, it was shown that intranasal administration of low concentrations of proteins leads to significant, rapid accumulation of protein in the optic nerve and in the eye, with only low levels reaching spinal cord and other brain regions. Thus, we examined whether intranasal inoculation with MHV at doses equivalent to those given intracranially could induce optic neuritis—inflammation, demyelination and loss of retinal ganglion cells (RGCs) in the optic nerve with or without inducing spinal cord demyelination. Four week old male C57BL/6J mice were inoculated intracranially with the recombinant demyelinating strain RSA59, or intranasally with RSA59 or the non-demyelinating strain RSMHV2 as control. One month post-inoculation, mice inoculated intracranially with RSA59 had significant myelin loss in both spinal cord and optic nerves, with significant loss of RGCs as well, consistent with prior studies. As expected, intranasal inoculation with RSA59 failed to induce demyelination in spinal cord; however, it also did not induce optic nerve demyelination. No acute inflammation was found, and no viral antigen was detected, in the optic nerve or retina 1 day after inoculation. Results confirm the neurotropic effects of RSA59 following intracranial inoculation, and suggest that direct infection with axonal transport of virus from brain to spinal cord and optic nerve is required to induce demyelinating disease. These studies suggest that MHV does not selectively concentrate in optic nerve and retina to sufficient levels to induce demyelination following intranasal inoculation. Intracranial inoculation should continue to be considered a preferred method for studies of MHV-induced optic neuritis and central nervous system (CNS) demyelinating disease.

Collaboration


Dive into the Kimberly Dine's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Reas S. Khan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zoe Fonseca-Kelly

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Mayssa Nassrallah

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Esteban Luna

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Helayna Brown

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Howard Wessel

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge