1
|
Chutia P, Tripathi SM, Jv A. The digitalization of psychopathology: 'TV sign' and 'Smartphone sign' as red flags for dementia. Neurocase 2025:1-5. [PMID: 39982201 DOI: 10.1080/13554794.2025.2467925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
This case series elucidates pathological signs for diagnosis in two patients with Dementia. The first case highlights the term 'Smartphone sign', a novel psychopathology uncovered based on the existing 'TV sign', a rare type of delusional misidentification syndrome (DMS). The second case had symptoms consistent with the 'TV sign'. The possible underlying cause of these signs was hypothesized based on psychopathology, brain region, sensory system, cognition, and environmental factors. Moreover, the treatment outcome in terms of cognition and behavior on low doses of Risperidone and Escitalopram shows promising results and paves the way for the treatment of other DMS.
Collapse
Affiliation(s)
- Porimita Chutia
- Department of Geriatric Mental Health, King George's Medical University, Lucknow, India
| | - Shailendra Mohan Tripathi
- Department of Geriatric Mental Health, King George's Medical University, Lucknow, India
- Institute of Medical Sciences, University of Aberdeen. Foresterhill, Aberdeen, UK
| | - Ashwin Jv
- Department of Geriatric Mental Health, King George's Medical University, Lucknow, India
| |
Collapse
|
2
|
Kaštelan S, Gverović Antunica A, Puzović V, Didović Pavičić A, Čanović S, Kovačević P, Vučemilović PAF, Konjevoda S. Non-Invasive Retinal Biomarkers for Early Diagnosis of Alzheimer's Disease. Biomedicines 2025; 13:283. [PMID: 40002697 PMCID: PMC11852429 DOI: 10.3390/biomedicines13020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the brain associated with ageing and is the most prevalent form of dementia, affecting an estimated 55 million people worldwide, with projections suggesting this number will exceed 150 million by 2050. With its increasing prevalence, AD represents a significant global health challenge with potentially serious social and economic consequences. Diagnosing AD is particularly challenging as it requires timely recognition. Currently, there is no effective therapy for AD; however, certain medications may help slow its progression. Existing diagnostic methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and biomarker analysis in cerebrospinal fluid tend to be expensive and invasive, making them impractical for widespread use. Consequently, research into non-invasive biomarkers that enable early detection and screening for AD is a crucial area of contemporary clinical investigation. One promising approach for the early diagnosis of AD may be retinal imaging. As an extension of the central nervous system, the retina offers a distinctive opportunity for non-invasive brain structure and function assessment. Considering their shared embryological origins and the vascular and immunological similarities between the eye and brain, alterations in the retina may indicate pathological changes in the brain, including those specifically related to AD. Studies suggest that structural and vascular changes in the retina, particularly within the neuronal network and blood vessels, may act as markers of cerebral changes caused by AD. These retinal alterations have the potential to act as biomarkers for early diagnosis. Since AD is typically diagnosed only after a significant neuronal loss has occurred, identifying early diagnostic markers could enable timely intervention and help prevent disease progression. Non-invasive retinal imaging techniques, such as optical coherence tomography (OCT) and OCT angiography, provide accessible methods for the early detection of changes linked to AD. This review article focuses on the potential of retinal imaging as a non-invasive biomarker for early diagnosis of AD. Investigating the ageing of the retina and its connections to neurodegenerative processes could significantly enhance the diagnosis, monitoring, and treatment of AD, paving the way for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Snježana Kaštelan
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Ophthalmology, Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | | | - Velibor Puzović
- Department of Pathology, General Hospital Dubrovnik, 20000 Dubrovnik, Croatia
| | | | - Samir Čanović
- Department of Ophthalmology, Zadar General Hospital, 23000 Zadar, Croatia
- Department of Health Studies, University of Zadar, 23000 Zadar, Croatia
| | - Petra Kovačević
- Department of Ophthalmology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | | | - Suzana Konjevoda
- Department of Ophthalmology, Zadar General Hospital, 23000 Zadar, Croatia
- Department of Health Studies, University of Zadar, 23000 Zadar, Croatia
| |
Collapse
|
3
|
Ahmed S, Son T, Ma G, Yao X. Polarization optical coherence tomography optoretinography: verifying light-induced photoreceptor outer segment shrinkage and subretinal space expansion. NEUROPHOTONICS 2025; 12:015005. [PMID: 39872019 PMCID: PMC11770343 DOI: 10.1117/1.nph.12.1.015005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
Significance Stimulus-evoked intrinsic optical signal (IOS) changes in retinal photoreceptors are critical for functional optoretinography (ORG). Optical coherence tomography (OCT), with its depth-resolved imaging capability, has been actively explored for IOS imaging of retinal photoreceptors. However, recent OCT studies have reported conflicting results regarding light-induced changes in the photoreceptor outer segments (OSs), with both elongation and shrinkage being observed. These discrepancies may stem from the difficulty in reliably identifying OS boundaries, particularly the inner segment/outer segment (IS/OS) junction and OS tip, as well as potential confusion with subretinal space dynamics. Gaining a better understanding of these light-induced OS changes is essential for accurate interpretation of ORG measurements and for optimizing IOS imaging systems to enhance sensitivity. Aim We aim to develop a method for the reliable identification of OS boundaries and to verify light-induced photoreceptor OS shrinkage and subretinal space expansion. Approach We employed a polarization-resolved full-field swept-source optical coherence tomography system capable of sequentially capturing parallel-polarization and cross-polarization OCT signals. The parallel-polarization mode is optimized to detect ballistically reflected photons from well-defined retinal boundaries, such as the IS/OS junction and the photoreceptor tips, whereas cross-polarization primarily captures multiply scattered photons. This differentiation enables parallel-polarization OCT to minimize the interference from scattered photons, enhancing the precision of OCT band quantification. Results Parallel-polarization OCT revealed photoreceptor OS shrinkage and subretinal space expansion in light conditions compared with dark conditions. Moreover, the overall outer retinal length appeared to swell under light. These observations were consistently confirmed in four healthy adult human subjects. Conclusions Parallel-polarization OCT provides a reliable method for identifying the IS/OS junction and OS tip, confirming light-induced photoreceptor OS shrinkage and subretinal space expansion.
Collapse
Affiliation(s)
- Shaiban Ahmed
- University of Illinois Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Taeyoon Son
- University of Illinois Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Guangying Ma
- University of Illinois Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Xincheng Yao
- University of Illinois Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
- University of Illinois Chicago, Department of Ophthalmology and Visual Sciences, Chicago, Illinois, United States
| |
Collapse
|
4
|
Molho W, Stiltner B, Raymond N, Kiely C, Trotti R, Harris C, Bannai D, Keshavan M, Silverstein S, Lizano P. Retinal electrophysiological alterations are associated with cognition in early course psychosis. Schizophr Res 2024; 274:137-141. [PMID: 39293251 DOI: 10.1016/j.schres.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024]
Abstract
Retinal electrophysiological alterations are implicated in psychosis, but their relationship with cognition in early course psychosis (ECP) is understudied. The Brief Assessment of Cognition (BAC) and flash electroretinography (fERG) were conducted in 24 controls (HC) and 27 ECP individuals. Partial Spearman correlations were performed between fERG and BAC. Lower Photopic-1b and Scotopic-3b amplitudes were identified in ECP vs. HCs. Correlations were significant (p<0.05) between BAC Composite score and a-wave S3a and S2a and b-wave S2b and S3b conditions. Thus, ECP was characterized by lower ERG responses, and lower rod/cone/bipolar cell responses were related to poorer cognition.
Collapse
Affiliation(s)
- Willa Molho
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Brendan Stiltner
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicolas Raymond
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Chelsea Kiely
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rebekah Trotti
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Caroline Harris
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Deepthi Bannai
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Steven Silverstein
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Division of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
5
|
Prasad M, Goodman D, Gutta S, Sheikh Z, Cabral HJ, Shunyakova J, Sanjiv N, Curley C, Yarala RR, Tsai L, Siegel NH, Chen X, Poulaki V, Alosco ML, Stein TD, Ness S, Subramanian ML. Associations Between Retinal Vascular Occlusions and Dementia. Healthcare (Basel) 2024; 12:2371. [PMID: 39684995 DOI: 10.3390/healthcare12232371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Retinal vascular occlusions, such as retinal vein occlusion (RVO) and retinal artery occlusion (RAO), are associated with cognitive impairment, including dementia. Our objective was to examine the odds of dementia among patients with retinal vascular occlusion. METHODS This cross-sectional study included 474 patients with retinal vascular occlusion and 948 patients without retinal vascular occlusion (comparison group). Patients in the comparison group were age- and sex-matched to those with vascular occlusion. Logistic regression was used to analyze the odds of all-cause dementia, vascular dementia, and Alzheimer's disease after adjusting for demographic, clinical, and ophthalmic covariates. Main outcome measures included the presence of all-cause dementia, vascular dementia, and Alzheimer's disease. RESULTS Patients with RVO (n = 413) had increased odds for all-cause dementia (odds ratio (OR) = 2.32; 95% confidence interval (CI): 1.44-3.75; p < 0.001) and vascular dementia (OR = 3.29; 95% CI: 1.41-7.68; p = 0.006) relative to the comparison group. Patients with central RVO (n = 192) (OR = 2.32; 95% CI: 1.19-4.54; p = 0.014) or branch RVO (n = 221) (OR = 2.68; 95% CI: 1.30-5.50; p = 0.007) had increased odds for all-cause dementia relative to the comparison group. Patients with RAO (n = 61) did not have increased odds of all-cause dementia (OR = 1.01; 95% CI: 0.32-3.26; p = 0.983), vascular dementia (OR = 1.54; 95% CI: 0.22-10.81; p = 0.663), or Alzheimer's disease (OR = 0.32; 95% CI: 0.05-2.20; p = 0.244). CONCLUSIONS A history of any RVO is associated with increased rates of all-cause dementia and vascular dementia independent of shared cardiovascular risk factors. These associations are not seen with a history of RAO, or between any subtype of vascular occlusions and Alzheimer's disease.
Collapse
Affiliation(s)
- Minali Prasad
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Deniz Goodman
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Sanhit Gutta
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Zahra Sheikh
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Howard J Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jenny Shunyakova
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Nayan Sanjiv
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Cameron Curley
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Rohun Reddy Yarala
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Lynna Tsai
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Nicole H Siegel
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Xuejing Chen
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Vasiliki Poulaki
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Thor D Stein
- VA Boston Healthcare System, Boston, MA 02130, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, Bedford, MA 01730, USA
| | - Steven Ness
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Manju L Subramanian
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
6
|
Curro KR, van Nispen RMA, den Braber A, van de Giessen EM, van de Kreeke JA, Tan HS, Visser PJ, Bouwman FH, Verbraak FD. Longitudinal Assessment of Retinal Microvasculature in Preclinical Alzheimer's Disease. Invest Ophthalmol Vis Sci 2024; 65:2. [PMID: 39361291 PMCID: PMC11451830 DOI: 10.1167/iovs.65.12.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Purpose To investigate if changes in vessel density (VD) and the foveal avascular zone (FAZ) occur in the preclinical phase of Alzheimer's disease (pAD) over time. Methods Optical coherence tomography angiography (OCTA) was used to image VD and FAZ at baseline and for a follow-up period of 2 years. Positron emission tomography (PET) was used to determine the amyloid beta (Aβ) status of participants. Results The VD and FAZ of 148 participants (54% female) were analyzed at baseline and follow-up (mean time between measurements, 2.24 ± 0.35 years). The mean age of the participants was 68.3 ± 6.0 years at baseline and 70.3 ± 5.9 years at follow-up. Participants were divided into three groups: control group, participants who had negative Aβ status at both measurements (Aβ-, n = 116); converter group, participants who transitioned from negative to positive between baseline and follow-up (Aβ-+, n = 18); and participants who were consistently positive at both visits (Aβ++, n = 14). The VD of both Aβ+ groups demonstrated non-significant increases over time in both macula and optic nerve head (ONH) regions. The Aβ- group was found to be significantly higher in both ONH and macular regions. The VD of the Aβ++ group was significantly higher in the macula inner and outer rings compared to the Aβ-+ and Aβ- groups. No significant change was found in FAZ values over time. Conclusions Alterations in VD seem to manifest already in pAD, exhibiting distinct variations between the ONH and macula. Further longitudinal studies with a longer follow-up design and known amyloid pathology should be undertaken to validate these observations.
Collapse
Affiliation(s)
- Katie R. Curro
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ruth M. A. van Nispen
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | - H. Stevie Tan
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pieter-Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Femke H. Bouwman
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frank D. Verbraak
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Blose BA, Silverstein SM, Stuart KV, Keane PA, Khawaja AP, Wagner SK. Association between polygenic risk for schizophrenia and retinal morphology: A cross-sectional analysis of the United Kingdom Biobank. Psychiatry Res 2024; 339:116106. [PMID: 39079374 DOI: 10.1016/j.psychres.2024.116106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024]
Abstract
We examined the relationship between genetic risk for schizophrenia (SZ), using polygenic risk scores (PRSs), and retinal morphological alterations. Retinal structural and vascular indices derived from optical coherence tomography (OCT) and color fundus photography (CFP) and PRSs for SZ were analyzed in N = 35,024 individuals from the prospective cohort study, United Kingdom Biobank (UKB). Results indicated that macular ganglion cell-inner plexiform layer (mGC-IPL) thickness was significantly inversely related to PRS for SZ, and this relationship was strongest within higher PRS quintiles and independent of potential confounders and age. PRS, however, was unrelated to retinal vascular characteristics, with the exception of venular tortuosity, and other retinal structural indices (macular retinal nerve fiber layer [mRNFL], inner nuclear layer [INL], cup-to-disc ratio [CDR]). Additionally, the association between greater PRS and reduced mGC-IPL thickness was only significant for participants in the 40-49 and 50-59 age groups, not those in the 60-69 age group. These findings suggest that mGC-IPL thinning is associated with a genetic predisposition to SZ and may reflect neurodevelopmental and/or neurodegenerative processes inherent to SZ. Retinal microvasculature alterations, however, may be secondary consequences of SZ and do not appear to be associated with a genetic predisposition to SZ.
Collapse
Affiliation(s)
- Brittany A Blose
- Department of Psychology, University of Rochester, Rochester, NY, United States; Department of Psychiatry, University of Rochester Medical Center, Rochester, New York, United States
| | - Steven M Silverstein
- Department of Psychiatry, University of Rochester Medical Center, Rochester, New York, United States; Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States; Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, United States; Center for Visual Science, University of Rochester, Rochester, New York, United States.
| | - Kelsey V Stuart
- NIHR Moorfields Biomedical Research Centre, London, United Kingdom; Institute of Ophthalmology, University College London, London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Pearse A Keane
- NIHR Moorfields Biomedical Research Centre, London, United Kingdom; Institute of Ophthalmology, University College London, London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Anthony P Khawaja
- NIHR Moorfields Biomedical Research Centre, London, United Kingdom; Institute of Ophthalmology, University College London, London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Siegfried K Wagner
- NIHR Moorfields Biomedical Research Centre, London, United Kingdom; Institute of Ophthalmology, University College London, London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
8
|
Wang XF, Xiang XH, Wei J, Zhang PB, Xu Q, Liu MH, Qu LQ, Wang XX, Yu L, Wu AG, Qing DL, Wu JM, Law BYK, Yu CL, Yong-Tang. Raddeanin A Protects the BRB Through Inhibiting Inflammation and Apoptosis in the Retina of Alzheimer's Disease. Neurochem Res 2024; 49:2197-2214. [PMID: 38834846 DOI: 10.1007/s11064-024-04145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/28/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Neuroinflammation and endothelial cell apoptosis are prominent features of blood-brain barrier (BBB) disruption, which have been described in Alzheimer's disease (AD) and can predict cognitive decline. Recent reports revealed vascular β-amyloid (Aβ) deposits, Muller cell degeneration and microglial dysfunction in the retina of AD patients. However, there has been no in-depth research on the roles of inflammation, retinal endothelial cell apoptosis, and blood-retinal barrier (BRB) damage in AD retinopathy. We found that Raddeanin A (RDA) could improve pathological and cognitive deficits in a mouse model of Alzheimer's disease by targeting β-amyloidosis, However, the effects of RDA on AD retinal function require further study. To clarify whether RDA inhibits inflammation and apoptosis and thus improves BRB function in AD-related retinopathy. In vitro we used Aβ-treated HRECs and MIO-M1 cells, and in vivo we used 3×Tg-AD mice to investigate the effect of RDA on BRB in AD-related retinopathy. We found that RDA could improve BRB function in AD-related retinopathy by inhibiting NLRP3-mediated inflammation and suppressing Wnt/β-catenin pathway-mediated apoptosis, which is expected to improve the pathological changes in AD-related retinopathy and the quality of life of AD patients.
Collapse
Affiliation(s)
- Xiao-Fang Wang
- Laboratory Animal Centre, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China
| | - Xiao-Hong Xiang
- Department of Ophthalmology in The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jing Wei
- Eye School and Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection of Chengdu University of TCM, Chengdu, China
| | - Peng-Bo Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 15651, China
| | - Qin Xu
- Department of Ophthalmology in The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Meng-Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li-Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xing-Xia Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Da-Lian Qing
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Jian-Ming Wu
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Betty Yuen-Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Chong-Lin Yu
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China.
| | - Yong-Tang
- Laboratory Animal Centre, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China.
| |
Collapse
|
9
|
Boff JM, Shrestha AP, Madireddy S, Viswaprakash N, Della Santina L, Vaithianathan T. The Interplay between Neurotransmitters and Calcium Dynamics in Retinal Synapses during Development, Health, and Disease. Int J Mol Sci 2024; 25:2226. [PMID: 38396913 PMCID: PMC10889697 DOI: 10.3390/ijms25042226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The intricate functionality of the vertebrate retina relies on the interplay between neurotransmitter activity and calcium (Ca2+) dynamics, offering important insights into developmental processes, physiological functioning, and disease progression. Neurotransmitters orchestrate cellular processes to shape the behavior of the retina under diverse circumstances. Despite research to elucidate the roles of individual neurotransmitters in the visual system, there remains a gap in our understanding of the holistic integration of their interplay with Ca2+ dynamics in the broader context of neuronal development, health, and disease. To address this gap, the present review explores the mechanisms used by the neurotransmitters glutamate, gamma-aminobutyric acid (GABA), glycine, dopamine, and acetylcholine (ACh) and their interplay with Ca2+ dynamics. This conceptual outline is intended to inform and guide future research, underpinning novel therapeutic avenues for retinal-associated disorders.
Collapse
Affiliation(s)
- Johane M. Boff
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.M.B.); (A.P.S.)
| | - Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.M.B.); (A.P.S.)
| | - Saivikram Madireddy
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Nilmini Viswaprakash
- Department of Medical Education, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | | | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.M.B.); (A.P.S.)
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
10
|
Reiss AB, Gulkarov S, Jacob B, Srivastava A, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Mitochondria in Alzheimer's Disease Pathogenesis. Life (Basel) 2024; 14:196. [PMID: 38398707 PMCID: PMC10890468 DOI: 10.3390/life14020196] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and incurable neurodegenerative disorder that primarily affects persons aged 65 years and above. It causes dementia with memory loss and deterioration in thinking and language skills. AD is characterized by specific pathology resulting from the accumulation in the brain of extracellular plaques of amyloid-β and intracellular tangles of phosphorylated tau. The importance of mitochondrial dysfunction in AD pathogenesis, while previously underrecognized, is now more and more appreciated. Mitochondria are an essential organelle involved in cellular bioenergetics and signaling pathways. Mitochondrial processes crucial for synaptic activity such as mitophagy, mitochondrial trafficking, mitochondrial fission, and mitochondrial fusion are dysregulated in the AD brain. Excess fission and fragmentation yield mitochondria with low energy production. Reduced glucose metabolism is also observed in the AD brain with a hypometabolic state, particularly in the temporo-parietal brain regions. This review addresses the multiple ways in which abnormal mitochondrial structure and function contribute to AD. Disruption of the electron transport chain and ATP production are particularly neurotoxic because brain cells have disproportionately high energy demands. In addition, oxidative stress, which is extremely damaging to nerve cells, rises dramatically with mitochondrial dyshomeostasis. Restoring mitochondrial health may be a viable approach to AD treatment.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Shelly Gulkarov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Benna Jacob
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Ankita Srivastava
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Mark M. Stecker
- The Fresno Institute of Neuroscience, Fresno, CA 93730, USA;
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
11
|
Suh A, Ong J, Kamran SA, Waisberg E, Paladugu P, Zaman N, Sarker P, Tavakkoli A, Lee AG. Retina Oculomics in Neurodegenerative Disease. Ann Biomed Eng 2023; 51:2708-2721. [PMID: 37855949 DOI: 10.1007/s10439-023-03365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/05/2023] [Indexed: 10/20/2023]
Abstract
Ophthalmic biomarkers have long played a critical role in diagnosing and managing ocular diseases. Oculomics has emerged as a field that utilizes ocular imaging biomarkers to provide insights into systemic diseases. Advances in diagnostic and imaging technologies including electroretinography, optical coherence tomography (OCT), confocal scanning laser ophthalmoscopy, fluorescence lifetime imaging ophthalmoscopy, and OCT angiography have revolutionized the ability to understand systemic diseases and even detect them earlier than clinical manifestations for earlier intervention. With the advent of increasingly large ophthalmic imaging datasets, machine learning models can be integrated into these ocular imaging biomarkers to provide further insights and prognostic predictions of neurodegenerative disease. In this manuscript, we review the use of ophthalmic imaging to provide insights into neurodegenerative diseases including Alzheimer Disease, Parkinson Disease, Amyotrophic Lateral Sclerosis, and Huntington Disease. We discuss recent advances in ophthalmic technology including eye-tracking technology and integration of artificial intelligence techniques to further provide insights into these neurodegenerative diseases. Ultimately, oculomics opens the opportunity to detect and monitor systemic diseases at a higher acuity. Thus, earlier detection of systemic diseases may allow for timely intervention for improving the quality of life in patients with neurodegenerative disease.
Collapse
Affiliation(s)
- Alex Suh
- Tulane University School of Medicine, New Orleans, LA, USA.
| | - Joshua Ong
- Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sharif Amit Kamran
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Ethan Waisberg
- University College Dublin School of Medicine, Belfield, Dublin, Ireland
| | - Phani Paladugu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nasif Zaman
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Prithul Sarker
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Andrew G Lee
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, 6560 Fannin St #450, Houston, TX, 77030, USA
- The Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
- Departments of Ophthalmology, Neurology and Neurosurgery, Weill Cornell Medicine, New York, NY, USA
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Texas A&M College of Medicine, Bryan, TX, USA
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
12
|
Boccaccini A, Cavaterra D, Carnevale C, Tanga L, Marini S, Bocedi A, Lacal PM, Manni G, Graziani G, Sbardella D, Tundo GR. Novel frontiers in neuroprotective therapies in glaucoma: Molecular and clinical aspects. Mol Aspects Med 2023; 94:101225. [PMID: 38000334 DOI: 10.1016/j.mam.2023.101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
In the last years, neuroprotective therapies have attracted the researcher interests as modern and challenging approach for the treatment of neurodegenerative diseases, aimed at protecting the nervous system from injuries. Glaucoma is a neurodegenerative disease characterized by progressive excavation of the optic nerve head, retinal axonal injury and corresponding vision loss that affects millions of people on a global scale. The molecular basis of the pathology is largely uncharacterized yet, and the therapeutic approaches available do not change the natural course of the disease. Therefore, in accordance with the therapeutic regimens proposed for other neurodegenerative diseases, a modern strategy to treat glaucoma includes prescription of drugs with neuroprotective activities. With respect to this, several preclinical and clinical investigations on a plethora of different drugs are currently ongoing. In this review, first, the conceptualization of the rationale for the adoption of neuroprotective strategies for retina is summarized. Second, the molecular aspects highlighting glaucoma as a neurodegenerative disease are reported. In conclusion, the molecular and pharmacological properties of most promising direct neuroprotective drugs used to delay glaucoma progression are examined, including: neurotrophic factors, NMDA receptor antagonists, the α2-adrenergic agonist, brimonidine, calcium channel blockers, antioxidant agents, nicotinamide and statins.
Collapse
Affiliation(s)
| | - Dario Cavaterra
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Tor Vergata, Via Della Ricerca Scientifica 1, 00133, Rome, Italy
| | | | | | - Stefano Marini
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Via Montpellier n. 1, 00133, Roma, Italy
| | - Alessio Bocedi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Tor Vergata, Via Della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Pedro M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Gianluca Manni
- IRCCS - Fondazione Bietti, Rome, Italy; Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Via Montpellier n. 1, 00133, Roma, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133, Rome, Italy
| | | | - Grazia Raffaella Tundo
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Via Montpellier n. 1, 00133, Roma, Italy.
| |
Collapse
|
13
|
Wang S, Jiang X, Peng W, Yang S, Pi R, Zhou S. Acrolein Induces Retinal Abnormalities of Alzheimer's Disease in Mice. Int J Mol Sci 2023; 24:13576. [PMID: 37686379 PMCID: PMC10487815 DOI: 10.3390/ijms241713576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
It is reported that retinal abnormities are related to Alzheimer's disease (AD) in patients and animal models. However, it is unclear whether the retinal abnormities appear in the mouse model of sporadic Alzheimer's disease (sAD) induced by acrolein. We investigated the alterations of retinal function and structure, the levels of β-amyloid (Aβ) and phosphorylated Tau (p-Tau) in the retina, and the changes in the retinal vascular system in this mouse model. We demonstrated that the levels of Aβ and p-Tau were increased in the retinas of mice from the acrolein groups. Subsequently, a decreased amplitudes of b-waves in the scotopic and photopic electroretinogram (ERG), decreased thicknesses of the retinal nerve fiber layer (RNFL) in the retina, and slight retinal venous beading were found in the mice induced by acrolein. We propose that sAD mice induced by acrolein showed abnormalities in the retina, which may provide a valuable reference for the study of the retina in sAD.
Collapse
Affiliation(s)
- Shuyi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiuying Jiang
- Department of Ophthalmology, Affiliated Foshan Hospital, Southern Medical University, Foshan 528000, China
| | - Weijia Peng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shuangjian Yang
- Guangdong Provincial Institute for Vision and Eye Research, Guangzhou 510060, China
| | - Rongbiao Pi
- School of Medicine, Sun Yat-sen University, Shenzhen 528406, China
| | - Shiyou Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
14
|
Latina V, Atlante A, Malerba F, La Regina F, Balzamino BO, Micera A, Pignataro A, Stigliano E, Cavallaro S, Calissano P, Amadoro G. The Cleavage-Specific Tau 12A12mAb Exerts an Anti-Amyloidogenic Action by Modulating the Endocytic and Bioenergetic Pathways in Alzheimer's Disease Mouse Model. Int J Mol Sci 2023; 24:ijms24119683. [PMID: 37298634 DOI: 10.3390/ijms24119683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Beyond deficits in hippocampal-dependent episodic memory, Alzheimer's Disease (AD) features sensory impairment in visual cognition consistent with extensive neuropathology in the retina. 12A12 is a monoclonal cleavage specific antibody (mAb) that in vivo selectively neutralizes the AD-relevant, harmful N-terminal 20-22 kDa tau fragment(s) (i.e., NH2htau) without affecting the full-length normal protein. When systemically injected into the Tg2576 mouse model overexpressing a mutant form of Amyloid Precursor Protein (APP), APPK670/671L linked to early onset familial AD, this conformation-specific tau mAb successfully reduces the NH2htau accumulating both in their brain and retina and, thus, markedly alleviates the phenotype-associated signs. By means of a combined biochemical and metabolic experimental approach, we report that 12A12mAb downregulates the steady state expression levels of APP and Beta-Secretase 1 (BACE-1) and, thus, limits the Amyloid beta (Aβ) production both in the hippocampus and retina from this AD animal model. The local, antibody-mediated anti-amyloidogenic action is paralleled in vivo by coordinated modulation of the endocytic (BIN1, RIN3) and bioenergetic (glycolysis and L-Lactate) pathways. These findings indicate for the first time that similar molecular and metabolic retino-cerebral pathways are modulated in a coordinated fashion in response to 12A12mAb treatment to tackle the neurosensorial Aβ accumulation in AD neurodegeneration.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| | - Francesca Malerba
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Bijorn Omar Balzamino
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Egidio Stigliano
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| |
Collapse
|
15
|
Samoilă L, Voștinaru O, Dinte E, Bodoki AE, Iacob BC, Bodoki E, Samoilă O. Topical Treatment for Retinal Degenerative Pathologies: A Systematic Review. Int J Mol Sci 2023; 24:ijms24098045. [PMID: 37175752 PMCID: PMC10178888 DOI: 10.3390/ijms24098045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/12/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The topical administration of medicines is the preferred route in ocular therapy, at least for the anterior segment of the eye. However, the eye's inherent functional and biological barriers all work against the active pharmaceutical ingredient (API) to efficiently reach the targeted retinal structures. The main objective of this article is to offer a systematic review of the scientific literature in recent years, focusing on the latest developments of topical treatment intended for retinal degenerative diseases. Database search returned 102 clinical studies, focused on topical treatment for age macular degeneration, macular edemas (in diabetic retinopathy, surgery related or in retinal dystrophies) or glaucoma. After the exclusion of low-powered studies and those combining vitreo-retinal surgery, 35 articles remained for analysis. Currently, the topical treatment of retinal degenerative diseases is limited by the difficulty to deliver effective drug concentrations to the posterior eye structures. However, in the case of drug classes like NSAIDs, the presence of certain molecular and metabolic features for specific representatives makes the topical administration currently feasible in several clinical contexts. For other drug classes, either a fine-tuning of the API's pharmacokinetic profile or the use of more advanced formulation strategies, such as rationally designed nanostructured drugs and vehicles, crystalline polymorphs or supramolecular complexes, could bring the much awaited breakthrough for a more predictable and controlled delivery towards the retinal structures and could eventually be employed in the future for the development of more effective ways of delivering drugs to the posterior eye, with the ultimate goal of improving their clinical efficacy.
Collapse
Affiliation(s)
- Lăcrămioara Samoilă
- Department of Physiology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400006 Cluj-Napoca, Romania
| | - Oliviu Voștinaru
- Department of Pharmacology, Physiology and Physiopathology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400349 Cluj-Napoca, Romania
| | - Elena Dinte
- Department of Pharmaceutical Technology and Biopharmaceutics, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andreea Elena Bodoki
- Department of General and Inorganic Chemistry, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania
| | - Bogdan-Cezar Iacob
- Department of Analytical Chemistry, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ede Bodoki
- Department of Analytical Chemistry, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ovidiu Samoilă
- Department of Ophthalmology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 400006 Cluj-Napoca, Romania
| |
Collapse
|
16
|
Latina V, De Introna M, Caligiuri C, Loviglio A, Florio R, La Regina F, Pignataro A, Ammassari-Teule M, Calissano P, Amadoro G. Immunotherapy with Cleavage-Specific 12A12mAb Reduces the Tau Cleavage in Visual Cortex and Improves Visuo-Spatial Recognition Memory in Tg2576 AD Mouse Model. Pharmaceutics 2023; 15:pharmaceutics15020509. [PMID: 36839831 PMCID: PMC9965010 DOI: 10.3390/pharmaceutics15020509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Tau-targeted immunotherapy is a promising approach for treatment of Alzheimer's disease (AD). Beyond cognitive decline, AD features visual deficits consistent with the manifestation of Amyloid β-protein (Aβ) plaques and neurofibrillary tangles (NFT) in the eyes and higher visual centers, both in animal models and affected subjects. We reported that 12A12-a monoclonal cleavage-specific antibody (mAb) which in vivo neutralizes the neurotoxic, N-terminal 20-22 kDa tau fragment(s)-significantly reduces the retinal accumulation in Tg(HuAPP695Swe)2576 mice of both tau and APP/Aβ pathologies correlated with local inflammation and synaptic deterioration. Here, we report the occurrence of N-terminal tau cleavage in the primary visual cortex (V1 area) and the beneficial effect of 12A12mAb treatment on phenotype-associated visuo-spatial deficits in this AD animal model. We found out that non-invasive administration of 12 A12mAb markedly reduced the pathological accumulation of both truncated tau and Aβ in the V1 area, correlated to significant improvement in visual recognition memory performance along with local increase in two direct readouts of cortical synaptic plasticity, including the dendritic spine density and the expression level of activity-regulated cytoskeleton protein Arc/Arg3.1. Translation of these findings to clinical therapeutic interventions could offer an innovative tau-directed opportunity to delay or halt the visual impairments occurring during AD progression.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Margherita De Introna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Chiara Caligiuri
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Alessia Loviglio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Rita Florio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Martine Ammassari-Teule
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Via Ercole Ramarini 32, 00015 Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-06-49255252
| |
Collapse
|
17
|
The Risk of Age-Related Macular Degeneration Is Reduced in Type 2 Diabetes Patients Who Use Metformin. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background: Whether metformin may reduce the risk of age-related macular degeneration (AMD) requires confirmation. This study compared the risk of AMD between ever users and never users of metformin matched on propensity score (PS) in Taiwanese patients with type 2 diabetes mellitus. Methods: We enrolled study subjects from Taiwan’s National Health Insurance. A total of 423,949 patients with new onset diabetes from 1999 to 2005 were identified. After excluding ineligible patients and enrolling only patients aged between 50 and 79 years, we created 13,303 pairs of ever users and never users of metformin matched on PS. The patients were followed from 1 January 2006 to 31 December 2011. We estimated hazard ratios by Cox regression. Results: AMD was newly diagnosed in 506 ever users and 639 never users. The respective incidence rates (per 100,000 person-years) were 778.72 and 1016.62. The hazard ratio (HR) and 95% confidence interval (CI) for ever versus never users was 0.756 (0.673–0.850). While ever users were categorized by tertiles of cumulative duration (<31.8, 31.8–63.9 and >63.9 months) and cumulative dose (<947.1, 947.1–2193.5 and >2193.5 g) of metformin, a dose–response pattern was observed. For the respective tertiles of cumulative duration, the HRs (95% CIs) were 1.131 (0.961–1.330), 0.821 (0.697–0.967) and 0.464 (0.384–0.561), while compared to never users. For the respective tertiles of cumulative dose, the HRs (95% CIs) were 1.131 (0.962–1.329), 0.739 (0.624–0.876) and 0.525 (0.438–0.629). A risk reduction among ever users was observed for all tertiles of defined daily dose but was most remarkable for the third tertile with a defined daily dose of >0.64. Subgroup analyses suggested that the benefit of metformin could be similarly observed among men and women and for age subgroups of 50–64 and 65–79 years. However, patients with diabetic retinopathy would not be significantly benefited and metformin did not seem to be preventive for exudative AMD. Conclusion: In general, metformin significantly reduces the risk of AMD.
Collapse
|
18
|
Hao X, Zhang W, Jiao B, Yang Q, Zhang X, Chen R, Wang X, Xiao X, Zhu Y, Liao W, Wang D, Shen L. Correlation between retinal structure and brain multimodal magnetic resonance imaging in patients with Alzheimer's disease. Front Aging Neurosci 2023; 15:1088829. [PMID: 36909943 PMCID: PMC9992546 DOI: 10.3389/fnagi.2023.1088829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Background The retina imaging and brain magnetic resonance imaging (MRI) can both reflect early changes in Alzheimer's disease (AD) and may serve as potential biomarker for early diagnosis, but their correlation and the internal mechanism of retinal structural changes remain unclear. This study aimed to explore the possible correlation between retinal structure and visual pathway, brain structure, intrinsic activity changes in AD patients, as well as to build a classification model to identify AD patients. Methods In the study, 49 AD patients and 48 healthy controls (HCs) were enrolled. Retinal images were obtained by optical coherence tomography (OCT). Multimodal MRI sequences of all subjects were collected. Spearman correlation analysis and multiple linear regression models were used to assess the correlation between OCT parameters and multimodal MRI findings. The diagnostic value of combination of retinal imaging and brain multimodal MRI was assessed by performing a receiver operating characteristic (ROC) curve. Results Compared with HCs, retinal thickness and multimodal MRI findings of AD patients were significantly altered (p < 0.05). Significant correlations were presented between the fractional anisotropy (FA) value of optic tract and mean retinal thickness, macular volume, macular ganglion cell layer (GCL) thickness, inner plexiform layer (IPL) thickness in AD patients (p < 0.01). The fractional amplitude of low frequency fluctuations (fALFF) value of primary visual cortex (V1) was correlated with temporal quadrant peripapillary retinal nerve fiber layer (pRNFL) thickness (p < 0.05). The model combining thickness of GCL and temporal quadrant pRNFL, volume of hippocampus and lateral geniculate nucleus, and age showed the best performance to identify AD patients [area under the curve (AUC) = 0.936, sensitivity = 89.1%, specificity = 87.0%]. Conclusion Our study demonstrated that retinal structure change was related to the loss of integrity of white matter fiber tracts in the visual pathway and the decreased LGN volume and functional metabolism of V1 in AD patients. Trans-synaptic axonal retrograde lesions may be the underlying mechanism. Combining retinal imaging and multimodal MRI may provide new insight into the mechanism of retinal structural changes in AD and may serve as new target for early auxiliary diagnosis of AD.
Collapse
Affiliation(s)
- Xiaoli Hao
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Radiology, Xiangya Hospital of Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Xinyue Zhang
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Ruiting Chen
- Department of Radiology, Xiangya Hospital of Central South University, Changsha, China
| | - Xin Wang
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital of Central South University, Changsha, China
| | - Dongcui Wang
- Department of Radiology, Xiangya Hospital of Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
19
|
Abstract
Schizophrenia is increasingly recognized as a systemic disease, characterized by dysregulation in multiple physiological systems (eg, neural, cardiovascular, endocrine). Many of these changes are observed as early as the first psychotic episode, and in people at high risk for the disorder. Expanding the search for biomarkers of schizophrenia beyond genes, blood, and brain may allow for inexpensive, noninvasive, and objective markers of diagnosis, phenotype, treatment response, and prognosis. Several anatomic and physiologic aspects of the eye have shown promise as biomarkers of brain health in a range of neurological disorders, and of heart, kidney, endocrine, and other impairments in other medical conditions. In schizophrenia, thinning and volume loss in retinal neural layers have been observed, and are associated with illness progression, brain volume loss, and cognitive impairment. Retinal microvascular changes have also been observed. Abnormal pupil responses and corneal nerve disintegration are related to aspects of brain function and structure in schizophrenia. In addition, studying the eye can inform about emerging cardiovascular, neuroinflammatory, and metabolic diseases in people with early psychosis, and about the causes of several of the visual changes observed in the disorder. Application of the methods of oculomics, or eye-based biomarkers of non-ophthalmological pathology, to the treatment and study of schizophrenia has the potential to provide tools for patient monitoring and data-driven prediction, as well as for clarifying pathophysiology and course of illness. Given their demonstrated utility in neuropsychiatry, we recommend greater adoption of these tools for schizophrenia research and patient care.
Collapse
Affiliation(s)
- Steven M Silverstein
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Joy J Choi
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Kyle M Green
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Rajeev S Ramchandran
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
20
|
Zibetti C. Deciphering the Retinal Epigenome during Development, Disease and Reprogramming: Advancements, Challenges and Perspectives. Cells 2022; 11:cells11050806. [PMID: 35269428 PMCID: PMC8908986 DOI: 10.3390/cells11050806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Retinal neurogenesis is driven by concerted actions of transcription factors, some of which are expressed in a continuum and across several cell subtypes throughout development. While seemingly redundant, many factors diversify their regulatory outcome on gene expression, by coordinating variations in chromatin landscapes to drive divergent retinal specification programs. Recent studies have furthered the understanding of the epigenetic contribution to the progression of age-related macular degeneration, a leading cause of blindness in the elderly. The knowledge of the epigenomic mechanisms that control the acquisition and stabilization of retinal cell fates and are evoked upon damage, holds the potential for the treatment of retinal degeneration. Herein, this review presents the state-of-the-art approaches to investigate the retinal epigenome during development, disease, and reprogramming. A pipeline is then reviewed to functionally interrogate the epigenetic and transcriptional networks underlying cell fate specification, relying on a truly unbiased screening of open chromatin states. The related work proposes an inferential model to identify gene regulatory networks, features the first footprinting analysis and the first tentative, systematic query of candidate pioneer factors in the retina ever conducted in any model organism, leading to the identification of previously uncharacterized master regulators of retinal cell identity, such as the nuclear factor I, NFI. This pipeline is virtually applicable to the study of genetic programs and candidate pioneer factors in any developmental context. Finally, challenges and limitations intrinsic to the current next-generation sequencing techniques are discussed, as well as recent advances in super-resolution imaging, enabling spatio-temporal resolution of the genome.
Collapse
Affiliation(s)
- Cristina Zibetti
- Department of Ophthalmology, Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, Building 36, 0455 Oslo, Norway
| |
Collapse
|
21
|
Merlo S, Spampinato SF, Lim D. Molecular Aspects of Cellular Dysfunction in Alzheimer's Disease: The Need for a Holistic View of the Early Pathogenesis. Biomolecules 2021; 11:biom11121807. [PMID: 34944450 PMCID: PMC8699369 DOI: 10.3390/biom11121807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, a socio-economic burden destined to worsen with increased population aging [...].
Collapse
Affiliation(s)
- Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy;
- Correspondence: (S.M.); (D.L.)
| | - Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy;
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
- Correspondence: (S.M.); (D.L.)
| |
Collapse
|