Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1932 2022-04-21 23:17:43 |
2 format correct Meta information modification 1932 2022-04-22 03:24:31 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mansilla, A.; , .; De La Villa, P.; Germain, F. Neurodegeneration and Death of Retinal Ganglion Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/22144 (accessed on 27 April 2024).
Mansilla A,  , De La Villa P, Germain F. Neurodegeneration and Death of Retinal Ganglion Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/22144. Accessed April 27, 2024.
Mansilla, Alicia, , Pedro De La Villa, Francisco Germain. "Neurodegeneration and Death of Retinal Ganglion Cells" Encyclopedia, https://encyclopedia.pub/entry/22144 (accessed April 27, 2024).
Mansilla, A., , ., De La Villa, P., & Germain, F. (2022, April 21). Neurodegeneration and Death of Retinal Ganglion Cells. In Encyclopedia. https://encyclopedia.pub/entry/22144
Mansilla, Alicia, et al. "Neurodegeneration and Death of Retinal Ganglion Cells." Encyclopedia. Web. 21 April, 2022.
Neurodegeneration and Death of Retinal Ganglion Cells
Edit

Retinal ganglion cells (RGC) transmit light and visual information to the brain via the long axons that form the optic nerve. RGCs are the measurable endpoints in current research into experimental therapies and diagnosis in multiple ocular pathologies, like glaucoma. RGC subtype classifications are based on morphological, functional, genetical, and immunohistochemical aspects.

retinal ganglion cells neurodegeneration

1. Retinal Images Techniques

Optical coherence tomography (OCT) can be temporal or spectral domain. This technique generates two-dimensional cross-sectional images from the optical backscattering of light and the time delay of the echo. If enough scans are available, it can generate three-dimensional images. In addition, its axial resolution is very high (1–15 μm) [1][2]. However, weak backscatter and low-contrast cell borders prevent direct visualization of individual RGC. The fundamental value of OCT is to provide high-resolution images of the thickness of the retinal nerve fiber layer (RNFL) [3][4][5][6]. Its performance has been improved by applying a Doppler development [7]; as well as with polarization-sensitive OCT, since ocular structures are capable of altering the polarization state of light, adding tissue-specific contrast to images, and allowing RGC axon densities to be measured [8]. Similarly, long-wavelength OCT, which reduces scatter [9], and swept-source OCT, which uses a tunable long wavelength can further increase resolution, represent significant improvements.
Confocal scanning laser ophthalmoscopy (cSLO) was established in 1980 [10], and is the most widely used retinal imaging technique. Its remarkable ability to adapt has been key to its survival as a diagnostic technique in glaucoma, providing high-resolution images with which subtle changes in the retina can be detected [11].
The cSLO methodology is based on the confocal microscopy technique, in which a laser scans the retina, and thanks to the pinhole, only light coming from certain depths is detected. In addition, as it is a narrow beam of light, no scattered light is produced; ensuring that only light from the desired focal plane is detected. In this way, a high lateral resolution is achieved, which allows the production of topographic images [10]. However, the axial resolution of conventional cSLO is poor. The key to improving the functionality of cSLO is the use of fluorescent markers. So, several research groups have used this combination to detect apoptotic RGCs in vivo using endogenous or exogenous markers and to be able to longitudinally monitor rodent RGCs in vivo [12][13][14][15][2][4]. In addition, the possibility of using various types of filters and laser wavelengths would allow double or multiple label detections. Limitations on its use only include pupil diameter and ocular opacities. However, the future of cSLO lies in adaptive optics (AO), in the ability to alter the scan amplitude speed, and in modifying the size of the pinhole.
One of these technical improvements in the ophthalmological field has been achieved through adaptive optics, since it allowed reducing optical aberrations [16]. Intrinsic optical aberrations were detected by analyzing the wavefront of light coming from the eye, and were corrected by electro-actuated deformable mirrors [17]. The main technical advantages of AO are improved lateral and axial resolution of retinal images, detection of smaller dots, and improved sensitivity to weak reflections. In combination with cSLO and OCT, it can improve fine detail resolution in vivo [16]. The association of AO with OCT increases the lateral resolution by five times compared to standard OCT, allowing direct visualization of individual cells without the need for an exogenous marker. This resolution allows individual nerve fiber bundles to be observed in humans in vivo [18][19]. Association with cSLO allows high-resolution in vivo visualization of fluorescently labeled rodent capillaries [20]; direct observation of cone-type photoreceptors through their intrinsic reflectance [21], and better resolution to analyze RNFL. The AO-SLO association was more accurate than the OCT [22]. All the above indicates that the future of cSLO lies in AO development. Although, in parallel, research is being carried out on small-aperture fast-scanning cSLO [23]. On the other hand, fluorescent labelers have an important role in further improving this technique.

2. Apoptosis Detection Techniques

The Detection of Retinal Apoptotic Cells (DARC) is a recent methodology that, through real-time non-invasive imaging technique applied to the detection of apoptotic RGC cells in vivo, has the potential to identify diseases in their early stages [12]. It consists of the injection of intravenous annexin-5 marked with fluorescence (ANX776). Annexin 5 has a high calcium-dependent affinity for negatively charged phosphatidylserine [24]. It has been observed that, during the early phase of apoptosis, phosphatildylserine is externalized in the outer membrane of neurons. [25]. Binding of phosphatidylserine to annexin 5 in the plasma membranes of apoptotic cells is detected by cSLO retinal examination. This examination allows counting the number of fluorescent spots representing each simple apoptotic RGC bound to annexin-5 to be calculated. During the cellular stress of early apoptosis, phosphatidyserine, normally intracellular, translocate to the outer plasma membrane, and is exposed to the outside of the cell, signaling it to be removed by phagocytic cells [26]. As this is one of the initial steps of the apoptotic cascade, it constitutes a much earlier marker than others, such as DNA fragmentation detected by the terminal labeling of deoxynucleotidyl transferase dUTP Nick (TUNEL) [12][27][17][19].
Detection of annexin-5 labeling by confocal scanning ophthalmoscopy (cSLO) focused on the RGC layer [28] allows for obtaining high-contrast fluorescent images that span between 35° and 55° of the retinal field. On the other hand, the ability of annexin 5 to cross the blood-brain barrier [29] allows the assessment of diseases in other areas of the nervous system, in addition to the retina.
This technique has proven useful in the investigation of neuroprotective therapeutic agents in glaucoma models, as well as in the relationship between glaucoma and Alzheimer’s disease [12][30][31] or the correlation between the number of apoptotic cells and axonal loss in RGC [32]. It has also served to establish the relationship between increased intraocular pressure and RGC apoptosis [30], and to verify in an intraocular hypertension model the reduction of apoptosis in vivo by the use of coenzyme Q10 [33]. Likewise, in the retina of diabetic mice, an increase in DARC counts was observed before vascular changes were perceived in the eye [34]; in a model of blue light exposure in rats this technique served to determine photoreceptors loss [35]; it made possible to verify the neuroprotective effect of brimonidine in glaucoma and the prevention of the formation of amyloid plaques [36], especially useful for early visualization of Alzheimer’s disease; it was used to determine the protective effect of modulating glutamatergic excitotoxicity [37]; or to demonstrate the therapeutic capacity of 2-Cl-IB-MECA to reduce apoptosis in vivo after partial transection of the optic nerve [38]; it was essential to detect the regeneration of injured axons after placing Schwann cells on the damaged optic nerve sheath [39]; in the same model, topical recombinant human nerve growth factor (rh-NGF) was found to be able to decrease apoptosis [40] by using this technique.
Especially interesting is the fact that this technique allows, through the retina, to observe the evolution of other diseases whose main symptoms affect other systems. Thus, the analysis of amyloid plaques in the retina and their relationship with apoptosis have shown a dose- and time-dependent relationship, so that, by preventing their formation or increasing their elimination, survival was increased [41]. In general, a close relationship has been observed between the retina and diseases in other parts of the nervous system, such as Alzheimer’s [42][43] or Parkinson’s diseases [44]. Therefore, the study of the retina seems to be a good way to control the evolution of these other diseases of the nervous system.

3. Caspase Activation Detection

Caspases are essential endoproteases in apoptotic and inflammatory processes. Caspase activation occurs through extrinsic or intrinsic signals. The extrinsic pathway is triggered by ligands that bind to extracellular death receptors, while the intrinsic pathway responds to intracellular stress signals such as hypoxia, DNA damage, reactive oxygen species, accumulation of misfolded proteins, and mitochondrial damage. Regardless of the trigger, the cascade begins with the activation of “starter” caspases capable of cleaving and activating “executer” caspases, which cleave DNA leading to cell death. Detection of these caspases is a reliable indicator of apoptosis. On the other hand, some data suggest that different types of neurons use different death messages. Thus, in the retina, caspase-1 plays an important role in photoreceptor death, while caspase-3 is important in the inner nuclear layer, and caspase-2 is the main caspase involved in neuron death in the retinal ganglion cell layer [45].
Another option to detect caspase activity is the use of apoptotic probes (CapQ) activated by caspases. These penetrate cells and mark those that are in apoptosis [27]. This technology consists of a cell-penetrating peptide conjugated to an effector caspase recognition sequence, joint to a pair of fluorophores. This set is activated by effector caspases in apoptotic cells, and its fluorescence is detected by cSLO. After intravitreal injection of the TcapQ488 probe, RGCs showing apoptosis in vivo were detected using cSLO in mouse retinal degeneration models [46]. However, this probe had minimal toxicity capable of activating the probe, even in the eyes of wild-type rodents, and therefore causing apoptosis.
The use of caspase inhibitors with fluorescence (FLIVO, fluorescence in vivo) allows the visualization of apoptosis in vivo and in vitro. These tracers injected into the circulation selectively accumulate in apoptotic cells. Being able to cross the blood-brain barrier, they can be used in the study of brain and eye neurodegenerative diseases, selectively targeting cells that undergo caspase-dependent apoptosis [47]. These methodologies have been used in animal models (in vitro and in vivo) and in the clinic to monitor the activity of Diabetic Retinopathy [48], glaucoma [49], retinitis pigmentosa [50], blue light-induced retinal damage, and AMD [51].
Luciferins are bioluminescent molecules that when activated by luciferases release energy by emitting light. Z-DEVD-aminoluciferin is a luciferin modified to be activated by specific caspases, which allows it to detect the activity of these caspases in vitro [52]. In this way, it serves as a marker of apoptosis. Apoptosis has also been detected in vivo in mouse models of tumor xenografts [53].

4. Detection of Changes in the Apoptotic Membrane

Certain imaging techniques to detect apoptotic cells also use low molecular weight (300 to 700 Da) amphipathic molecules that selectively cross the apoptotic plasma membrane and accumulate in its cytoplasm as the Aposense family of compounds [54][55]. The anchoring is made to the hydrophobic (lipid) region of the cell membrane, passing into the cell interior, unlike what happens in non-apoptotic cells, in which the hydrophilic region blocks their entry into the cytoplasm. Examples of these compounds are those containing the dansyl group, like N,N′-didansyl-L-cystine, NST-732, and NST-729; or those containing an alkyl-malonate molecule, such as ML-9 and ML-10 [54].In apoptosis, the accumulation of these molecules in the cytoplasm, exposure to phosphatidylserine, activation of caspases, and loss of mitochondrial membrane potentials have been observed. These compounds may be intrinsically fluorescent or may be labeled with a radioactive moiety. These molecules have demonstrated their usefulness in the field of experimentation in models of Alzheimer’s disease, amyotrophic lateral sclerosis [56], melanomas [57], chemotherapy-induced enteropathy [58], and models of reperfusion-induced damage [55]. In clinical practice, a radiolabeled version of ML-10 has been used to monitor the response of brain metastases to radiotherapy [59]. Although these molecules can cross the blood-brain barrier, and therefore could be used in neurodegenerative conditions such as Alzheimer’s, Parkinson’s, and glaucoma, their toxicity could be a problem.
Glaucoma ocular biomarkers are endogenous biochemical, physiological, and anatomical indicators associated with specific pathological states [60][61]. They provide an objective measure to detect the disease early and monitor therapeutic efficacy. The optimal biomarker must be specific, sensitive, and reproducible, as well as inexpensive and non-invasive. A glaucoma biomarker should indicate the rate of RGC loss and the number of remaining or apoptotic RGCs with high sensitivity. Recent advances in fluorescent technology have improved the ability to identify individual RGCs undergoing apoptosis. These specific markings will be valuable both in experimental models and in the clinic.

References

  1. Costa, R.A.; Skaf, M.; Melo, L.A.S.; Calucci, D.; Cardillo, J.A.; Castro, J.C.; Huang, D.; Wojtkowski, M. Retinal Assessment Using Optical Coherence Tomography. Prog. Retin. Eye Res. 2006, 25, 325–353.
  2. Schuman, J.S. Spectral Domain Optical Coherence Tomography for Glaucoma (an AOS Thesis). Trans. Am. Ophthalmol. Soc. 2008, 106, 426–458.
  3. Leung, C.K.; Lindsey, J.D.; Crowston, J.G.; Lijia, C.; Chiang, S.; Weinreb, R.N. Longitudinal Profile of Retinal Ganglion Cell Damage after Optic Nerve Crush with Blue-Light Confocal Scanning Laser Ophthalmoscopy. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4898–4902.
  4. Petzold, A.; de Boer, J.F.; Schippling, S.; Vermersch, P.; Kardon, R.; Green, A.; Calabresi, P.A.; Polman, C. Optical Coherence Tomography in Multiple Sclerosis: A Systematic Review and Meta-Analysis. Lancet Neurol. 2010, 9, 921–932.
  5. Bussel, I.I.; Wollstein, G.; Schuman, J.S. OCT for Glaucoma Diagnosis, Screening and Detection of Glaucoma Progression. Br. J. Ophthalmol. 2014, 98 (Suppl. S2), ii15–ii19.
  6. Werkmeister, R.M.; Dragostinoff, N.; Pircher, M.; Götzinger, E.; Hitzenberger, C.K.; Leitgeb, R.A.; Schmetterer, L. Bidirectional Doppler Fourier-Domain Optical Coherence Tomography for Measurement of Absolute Flow Velocities in Human Retinal Vessels. Opt. Lett. 2008, 33, 2967–2969.
  7. Pircher, M.; Hitzenberger, C.K.; Schmidt-Erfurth, U. Polarization Sensitive Optical Coherence Tomography in the Human Eye. Prog. Retin. Eye Res. 2011, 30, 431–451.
  8. Chauhan, B.C. Confocal Scanning Laser Tomography. Can. J. Ophthalmol. 1996, 31, 152–156.
  9. Cordeiro, M.F.; Guo, L.; Coxon, K.M.; Duggan, J.; Nizari, S.; Normando, E.M.; Sensi, S.L.; Sillito, A.M.; Fitzke, F.W.; Salt, T.E.; et al. Imaging Multiple Phases of Neurodegeneration: A Novel Approach to Assessing Cell Death in Vivo. Cell Death Dis. 2010, 1, e3.
  10. Keane, P.A.; Ruiz-Garcia, H.; Sadda, S.R. Clinical Applications of Long-Wavelength (1000-Nm) Optical Coherence Tomography. Ophthalmic Surg. Lasers Imaging Off. J. Int. Soc. Imaging Eye 2011, 42 (Suppl. S4), S67–S74.
  11. Webb, R.H.; Hughes, G.W.; Delori, F.C. Confocal Scanning Laser Ophthalmoscope. Appl. Opt. 1987, 26, 1492–1499.
  12. Gray, D.C.; Merigan, W.; Wolfing, J.I.; Gee, B.P.; Porter, J.; Dubra, A.; Twietmeyer, T.H.; Ahamd, K.; Tumbar, R.; Reinholz, F.; et al. In Vivo Fluorescence Imaging of Primate Retinal Ganglion Cells and Retinal Pigment Epithelial Cells. Opt. Express 2006, 14, 7144–7158.
  13. Briggman, K.L.; Helmstaedter, M.; Denk, W. Wiring Specificity in the Direction-Selectivity Circuit of the Retina. Nature 2011, 471, 183–188.
  14. Nuschke, A.C.; Farrell, S.R.; Levesque, J.M.; Chauhan, B.C. Assessment of Retinal Ganglion Cell Damage in Glaucomatous Optic Neuropathy: Axon Transport, Injury and Soma Loss. Exp. Eye Res. 2015, 141, 111–124.
  15. Krieger, B.; Qiao, M.; Rousso, D.L.; Sanes, J.R.; Meister, M. Four Alpha Ganglion Cell Types in Mouse Retina: Function, Structure, and Molecular Signatures. PLoS ONE 2017, 12, e0180091.
  16. Miller, D.T.; Kocaoglu, O.P.; Wang, Q.; Lee, S. Adaptive Optics and the Eye (Super Resolution OCT). Eye Lond. Engl. 2011, 25, 321–330.
  17. Liang, J.; Grimm, B.; Goelz, S.; Bille, J.F. Objective Measurement of Wave Aberrations of the Human Eye with the Use of a Hartmann-Shack Wave-Front Sensor. J. Opt. Soc. Am. A Opt. Image Sci. Vis. 1994, 11, 1949–1957.
  18. Chen, T.C.; Cense, B.; Pierce, M.C.; Nassif, N.; Park, B.H.; Yun, S.H.; White, B.R.; Bouma, B.E.; Tearney, G.J.; de Boer, J.F. Spectral Domain Optical Coherence Tomography: Ultra-High Speed, Ultra-High Resolution Ophthalmic Imaging. Arch. Ophthalmol. Chic. Ill 1960 2005, 123, 1715–1720.
  19. Truong, S.N.; Alam, S.; Zawadzki, R.J.; Choi, S.S.; Telander, D.G.; Park, S.S.; Werner, J.S.; Morse, L.S. High Resolution Fourier-Domain Optical Coherence Tomography of Retinal Angiomatous Proliferation. Retina Phila. Pa 2007, 27, 915–925.
  20. Biss, D.P.; Sumorok, D.; Burns, S.A.; Webb, R.H.; Zhou, Y.; Bifano, T.G.; Côté, D.; Veilleux, I.; Zamiri, P.; Lin, C.P. In Vivo Fluorescent Imaging of the Mouse Retina Using Adaptive Optics. Opt. Lett. 2007, 32, 659–661.
  21. Williams, D.R. Imaging Single Cells in the Living Retina. Vision Res. 2011, 51, 1379–1396.
  22. Chen, M.F.; Chui, T.Y.P.; Alhadeff, P.; Rosen, R.B.; Ritch, R.; Dubra, A.; Hood, D.C. Adaptive Optics Imaging of Healthy and Abnormal Regions of Retinal Nerve Fiber Bundles of Patients with Glaucoma. Investig. Ophthalmol. Vis. Sci. 2015, 56, 674–681.
  23. Cilkova, M.; Matlach, J.; Chopra, R.; Rider, A.; Shah, N.; Mulholland, P.; Dakin, S.C.; Tufail, A.; Anderson, R.S. Repeatability and Inter-Observer Variability of in Vivo Retinal Cone Imaging Using a Modified Heidelberg Retinal Angiography (HRA2) in Normal Subjects. Investig. Ophthalmol. Vis. Sci. 2015, 56, 4921.
  24. Meers, P.; Mealy, T. Calcium-Dependent Annexin V Binding to Phospholipids: Stoichiometry, Specificity, and the Role of Negative Charge. Biochemistry 1993, 32, 11711–11721.
  25. Yap, T.E.; Davis, B.M.; Guo, L.; Normando, E.M.; Cordeiro, M.F. Annexins in Glaucoma. Int. J. Mol. Sci. 2018, 19, 1218.
  26. Fadok, V.A.; Voelker, D.R.; Campbell, P.A.; Cohen, J.J.; Bratton, D.L.; Henson, P.M. Exposure of Phosphatidylserine on the Surface of Apoptotic Lymphocytes Triggers Specific Recognition and Removal by Macrophages. J. Immunol. Baltim. Md 1950 1992, 148, 2207–2216.
  27. Qiu, X.; Johnson, J.R.; Wilson, B.S.; Gammon, S.T.; Piwnica-Worms, D.; Barnett, E.M. Single-Cell Resolution Imaging of Retinal Ganglion Cell Apoptosis in Vivo Using a Cell-Penetrating Caspase-Activatable Peptide Probe. PLoS ONE 2014, 9, e88855.
  28. Adanja, I.; Debeir, O.; Mégalizzi, V.; Kiss, R.; Warzée, N.; Decaestecker, C. Automated Tracking of Unmarked Cells Migrating in Three-Dimensional Matrices Applied to Anti-Cancer Drug Screening. Exp. Cell Res. 2010, 316, 181–193.
  29. D’Arceuil, H.; Rhine, W.; de Crespigny, A.; Yenari, M.; Tait, J.F.; Strauss, W.H.; Engelhorn, T.; Kastrup, A.; Moseley, M.; Blankenberg, F.G. 99mTc Annexin V Imaging of Neonatal Hypoxic Brain Injury. Stroke 2000, 31, 2692–2700.
  30. Guo, L.; Moss, S.E.; Alexander, R.A.; Ali, R.R.; Fitzke, F.W.; Cordeiro, M.F. Retinal Ganglion Cell Apoptosis in Glaucoma Is Related to Intraocular Pressure and IOP-Induced Effects on Extracellular Matrix. Investig. Ophthalmol. Vis. Sci. 2005, 46, 175–182.
  31. Guo, L.; Duggan, J.; Cordeiro, M.F. Alzheimer’s Disease and Retinal Neurodegeneration. Curr. Alzheimer Res. 2010, 7, 3–14.
  32. Baltmr, A.; Duggan, J.; Nizari, S.; Salt, T.; Cordeiro, M. Neuroprotection in Glaucoma—Is There a Future Role? Exp. Eye Res. 2010, 91, 554–566.
  33. Davis, B.M.; Tian, K.; Pahlitzsch, M.; Brenton, J.; Ravindran, N.; Butt, G.; Malaguarnera, G.; Normando, E.M.; Guo, L.; Cordeiro, M.F. Topical Coenzyme Q10 Demonstrates Mitochondrial-Mediated Neuroprotection in a Rodent Model of Ocular Hypertension. Mitochondrion 2017, 36, 114–123.
  34. Borrie, S.C.; Cheung, W.; Guo, L.; Barber, A.J.; Singh, R.S.J.; Gardner, T.W.; Cordeiro, M.F. Diabetic Retinal Neurodegeneration: In Vivo Imaging of Retinal Ganglion Cell Apoptosis in the Ins2Akita/J Mouse. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4924.
  35. Schmitz-Valckenberg, S.; Guo, L.; Cheung, W.; Moss, S.E.; Fitzke, F.W.; Cordeiro, M.F. . Ophthalmol. Z. Dtsch. Ophthalmol. Ges. 2010, 107, 22–29.
  36. Nizari, S.; Guo, L.; Davis, B.M.; Normando, E.M.; Galvao, J.; Turner, L.A.; Bizrah, M.; Dehabadi, M.; Tian, K.; Cordeiro, M.F. Non-Amyloidogenic Effects of A2 Adrenergic Agonists: Implications for Brimonidine-Mediated Neuroprotection. Cell Death Dis. 2016, 7, e2514.
  37. Guo, L.; Salt, T.E.; Maass, A.; Luong, V.; Moss, S.E.; Fitzke, F.W.; Cordeiro, M.F. Assessment of Neuroprotective Effects of Glutamate Modulation on Glaucoma-Related Retinal Ganglion Cell Apoptosis in Vivo. Investig. Ophthalmol. Vis. Sci. 2006, 47, 626–633.
  38. Galvao, J.; Elvas, F.; Martins, T.; Cordeiro, M.F.; Ambrósio, A.F.; Santiago, A.R. Adenosine A3 Receptor Activation Is Neuroprotective against Retinal Neurodegeneration. Exp. Eye Res. 2015, 140, 65–74.
  39. Guo, L.; Davis, B.; Nizari, S.; Normando, E.M.; Shi, H.; Galvao, J.; Turner, L.; Shi, J.; Clements, M.; Parrinello, S.; et al. Direct Optic Nerve Sheath (DONS) Application of Schwann Cells Prolongs Retinal Ganglion Cell Survival in Vivo. Cell Death Dis. 2014, 5, e1460.
  40. Guo, L.; Davis, B.M.; Ravindran, N.; Galvao, J.; Kapoor, N.; Haamedi, N.; Shamsher, E.; Luong, V.; Fico, E.; Cordeiro, M.F. Topical Recombinant Human Nerve Growth Factor (Rh-NGF) Is Neuroprotective to Retinal Ganglion Cells by Targeting Secondary Degeneration. Sci. Rep. 2020, 10, 3375.
  41. Guo, L.; Salt, T.E.; Luong, V.; Wood, N.; Cheung, W.; Maass, A.; Ferrari, G.; Russo-Marie, F.; Sillito, A.M.; Cheetham, M.E.; et al. Targeting Amyloid-Beta in Glaucoma Treatment. Proc. Natl. Acad. Sci. USA 2007, 104, 13444–13449.
  42. Salt, T.E.; Nizari, S.; Cordeiro, M.F.; Russ, H.; Danysz, W. Effect of the Aβ Aggregation Modulator MRZ-99030 on Retinal Damage in an Animal Model of Glaucoma. Neurotox. Res. 2014, 26, 440–446.
  43. Sánchez-López, E.; Egea, M.A.; Davis, B.M.; Guo, L.; Espina, M.; Silva, A.M.; Calpena, A.C.; Souto, E.M.B.; Ravindran, N.; Ettcheto, M.; et al. Memantine-Loaded PEGylated Biodegradable Nanoparticles for the Treatment of Glaucoma. Small Weinh. Bergstr. Ger. 2018, 14, 1701808.
  44. Normando, E.M.; Davis, B.M.; De Groef, L.; Nizari, S.; Turner, L.A.; Ravindran, N.; Pahlitzsch, M.; Brenton, J.; Malaguarnera, G.; Guo, L.; et al. The Retina as an Early Biomarker of Neurodegeneration in a Rotenone-Induced Model of Parkinson’s Disease: Evidence for a Neuroprotective Effect of Rosiglitazone in the Eye and Brain. Acta Neuropathol. Commun. 2016, 4, 86.
  45. Yoshimura, N. . Nippon Ganka Gakkai Zasshi 2001, 105, 884–902.
  46. Nickerson, J.M.; Getz, S.E.; Sellers, J.T.; Chrenek, M.A.; Goodman, P.; Bernal, C.J.; Boatright, J.H. DNA Delivery in Adult Mouse Eyes: An Update with Corneal Outcomes. Methods Mol. Biol. Clifton NJ 2014, 1121, 165–177.
  47. Griffin, R.J.; Williams, B.W.; Bischof, J.C.; Olin, M.; Johnson, G.L.; Lee, B.W. Use of a Fluorescently Labeled Poly-Caspase Inhibitor for in Vivo Detection of Apoptosis Related to Vascular-Targeting Agent Arsenic Trioxide for Cancer Therapy. Technol. Cancer Res. Treat. 2007, 6, 651–654.
  48. Perrone, L.; Devi, T.S.; Hosoya, K.-I.; Terasaki, T.; Singh, L.P. Inhibition of TXNIP Expression in Vivo Blocks Early Pathologies of Diabetic Retinopathy. Cell Death Dis. 2010, 1, e65.
  49. Bosco, A.; Crish, S.D.; Steele, M.R.; Romero, C.O.; Inman, D.M.; Horner, P.J.; Calkins, D.J.; Vetter, M.L. Early Reduction of Microglia Activation by Irradiation in a Model of Chronic Glaucoma. PLoS ONE 2012, 7, e43602.
  50. Subramani, M.; Murugeswari, P.; Dhamodaran, K.; Chevour, P.; Gunasekaran, S.; Kumar, R.S.; Jayadev, C.; Shetty, R.; Begum, N.; Das, D. Short Pulse of Clinical Concentration of Bevacizumab Modulates Human Retinal Pigment Epithelial Functionality. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1140–1152.
  51. Dib, B.; Lin, H.; Maidana, D.E.; Tian, B.; Miller, J.B.; Bouzika, P.; Miller, J.W.; Vavvas, D.G. Mitochondrial DNA Has a Pro-Inflammatory Role in AMD. Biochim. Biophys. Acta 2015, 1853, 2897–2906.
  52. O’Brien, M.A.; Daily, W.J.; Hesselberth, P.E.; Moravec, R.A.; Scurria, M.A.; Klaubert, D.H.; Bulleit, R.F.; Wood, K.V. Homogeneous, Bioluminescent Protease Assays: Caspase-3 as a Model. J. Biomol. Screen. 2005, 10, 137–148.
  53. Scabini, M.; Stellari, F.; Cappella, P.; Rizzitano, S.; Texido, G.; Pesenti, E. In Vivo Imaging of Early Stage Apoptosis by Measuring Real-Time Caspase-3/7 Activation. Apoptosis Int. J. Program. Cell Death 2011, 16, 198–207.
  54. Grimberg, H.; Levin, G.; Shirvan, A.; Cohen, A.; Yogev-Falach, M.; Reshef, A.; Ziv, I. Monitoring of Tumor Response to Chemotherapy in Vivo by a Novel Small-Molecule Detector of Apoptosis. Apoptosis Int. J. Program. Cell Death 2009, 14, 257–267.
  55. Damianovich, M.; Ziv, I.; Heyman, S.N.; Rosen, S.; Shina, A.; Kidron, D.; Aloya, T.; Grimberg, H.; Levin, G.; Reshef, A.; et al. ApoSense: A Novel Technology for Functional Molecular Imaging of Cell Death in Models of Acute Renal Tubular Necrosis. Eur. J. Nucl. Med. Mol. Imaging 2006, 33, 281–291.
  56. Shirvan, A.; Reshef, A.; Yogev-Falach, M.; Ziv, I. Molecular Imaging of Neurodegeneration by a Novel Cross-Disease Biomarker. Exp. Neurol. 2009, 219, 274–283.
  57. Cohen, A.; Ziv, I.; Aloya, T.; Levin, G.; Kidron, D.; Grimberg, H.; Reshef, A.; Shirvan, A. Monitoring of Chemotherapy-Induced Cell Death in Melanoma Tumors by N,N′-Didansyl-L-Cystine. Technol. Cancer Res. Treat. 2007, 6, 221–234.
  58. Levin, G.; Shirvan, A.; Grimberg, H.; Reshef, A.; Yogev-Falach, M.; Cohen, A.; Ziv, I. Novel Fluorescence Molecular Imaging of Chemotherapy-Induced Intestinal Apoptosis. J. Biomed. Opt. 2009, 14, 054019.
  59. Höglund, J.; Shirvan, A.; Antoni, G.; Gustavsson, S.-Å.; Långström, B.; Ringheim, A.; Sörensen, J.; Ben-Ami, M.; Ziv, I. 18F-ML-10, a PET Tracer for Apoptosis: First Human Study. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 2011, 52, 720–725.
  60. Heaton, G.R.; Davis, B.M.; Turner, L.A.; Cordeiro, M.F. Ocular Biomarkers of Alzheimer’s Disease. Cent. Nerv. Syst. Agents Med. Chem. 2015, 15, 117–125.
  61. Bhattacharya, S.; Lee, R.; Grus, F. Molecular Biomarkers in Glaucoma. Investig. Ophthalmol. Vis. Sci. 2013, 54, 121–131.
More
Information
Subjects: Ophthalmology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 420
Revisions: 2 times (View History)
Update Date: 22 Apr 2022
1000/1000