You are on page 1of 14

Biomedicine & Pharmacotherapy 149 (2022) 112868

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Insulin like growth factor-1 works synergistically with dopamine to


attenuate diabetic retinopathy by downregulating vascular endothelial
growth factor
Shikha Upreti a, Seema Sen b, Tapas Chandra Nag c, Madhumita P. Ghosh a, *
a
Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida 201313, India
b
Department of Ocular Pathology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
c
Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India

A R T I C L E I N F O A B S T R A C T

Keywords: Aim: Levels of Insulin-like growth factor-1 (IGF-1), a proangiogenic growth factor is elevated and dopamine
Diabetic retinopathy downregulated in proliferative diabetic retinopathy (PDR). This study aims to investigate whether IGF-1 with
Dopamine dopamine can together modulate vascular endothelial growth factor (VEGF) to prevent proliferative diabetic
Insulin-like growth factor-1
retinopathy while also attenuating angiogenic effects of IGF-1.
Vascular dysfunction
Vascular endothelial growth factor
Methods: Effect of combination of levodopa L-Dopa with IGF-1 was tested on normal retinal pigment epithelium
CAM assay cells (ARPE-19) and human umbilical vein endothelial cells (HUVEC), followed by tube formation. Invivo
analysis of anti-angiogenic potential assessed by chick chorioallantoic membrane (CAM) assay. Diabetes in­
duction in wistar rats at two time points, 12 and 16 weeks, treated with L-Dopa+IGF-1 and analysed for
morphological variations, serum and tissue dopamine levels, gene expression by real-time PCR and western blot
assay.
Results: L-Dopa+IGF-1 on ARPE-19 cells caused no toxicity and worked synergistically. Reduced number of
vessels observed. Significant improvement in inner retina thickness (*p < 0.05) was observed when L-Dopa was
given alone and/or with IGF-1. Dopamine levels improved significantly in both serum and tissue (*p < 0.05).
Levels of VEGF and IGF-1 receptors reduced significantly in 12 weeks. Western studies suggest that L-Dopa+IGF-
1 modulates its effects via Akt/ERK dependent pathway.
Conclusion: First ever report on synergistic effect of L-Dopa+IGF-1 in a rat model of diabetic retinopathy. Even
though the effect of L-Dopa in combination with IGF-1 is comparable to levels of L-Dopa alone, this study
presents an interesting finding of neuroprotective function of IGF-1, which has been studied in disease models of
Parkinson’s but not diabetes.

1. Introduction positive retinal neurons within 6 months and dysfunction of inner retina
(IR) elements [2,4–7]. Various studies have found that L-Dopa admin­
Anatomical changes in neural retina following long-term prevalence istration at early stage of diabetic retinopathy may restore dopamine
of diabetes often lead to gradual deterioration of vision. Biosynthesis of levels, thereby improving retinal function resulting from vascular de­
dopamine, a key retinal neuromodulator contributes to normal vision fects that characterize clinically recognized diabetic retinopathy [8–12].
through sympathetic neurotransmission via regulation of its receptors Hence, dopamine supplementation has been found to restore retinal
and retinal pathways [1,2]. Chronic hyperglycemia resulting from dia­ function and is the most prospective regulator of diabetes induced
betes alters neuronal function by reducing synthesis of dopamine in metergasis of retina [3].
retina and causes its frequent efflux, modifying the dopaminergic com­ Increased levels of IGF-1 in vitreous have been correlated with
munications in retina [3]. Aberrations in retinal dopamine levels in as severity of diabetic retinal neovascularization [6,13–15]. Another
early as 4 weeks of diabetes results in loss of tyrosine hydroxylase crucial consequence of diabetic retinopathy is appearance of vascular

* Correspondence to: Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar
Pradesh, Room no. 322, J-3 block, Noida 201313, India.
E-mail addresses: shikha05upreti@gmail.com (S. Upreti), ssenop@rediffmail.com (S. Sen), tapas_nag@yahoo.com (T.C. Nag), mpghosh@amity.edu (M.P. Ghosh).

https://doi.org/10.1016/j.biopha.2022.112868
Received 5 December 2021; Received in revised form 16 March 2022; Accepted 23 March 2022
Available online 1 April 2022
0753-3322/© 2022 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

lesions in retina and onset of neovascularization. Although, various Dopa (0–50 µM) and IGF-1 (26 nM) alone or co-treatment with
studies have suggested that higher serum IGF-1 levels pose a risk to­ high glucose (HG) at 50 mM [25]. The dosing solutions of L-Dopa
wards development of severe diabetic retinopathy, on the contrary, and IGF-1 were prepared from 200 mM and 100 µg/ml stocks,
there are other studies that show no association between serum IGF-1 respectively in DMEM F-12 medium with low serum. After 24 h of
levels with development or progression of diabetic retinopathy treatment, 3-(4, 5-dimethylthiazol-2-yl)− 2, 5-diphenyl tetrazo­
[16–18]. Importantly, restricting IGF-1-induced signaling cascades lium bromide (MTT) was added to each well, incubated for 4 h,
effectively reduces retinal VEGF levels and blood-retinal barrier (BRB) crystals were dissolved with dimethyl sulfoxide (DMSO) and
breakdown [18]. IGF-1 controls striatal dopamine levels, local doap­ absorbance measured at 570 nm.
mine release in the midbrain, and dopamine neuron firing, ultimately (b) The IC50 values of IGF-1 and L-Dopa was estimated on ARPE-19
controlling dopamine dependent behaviors[19]. Also known to protect cells and degree of drug interaction was assessed by combination
and promote the survival of dopaminergic neurons invitro, chronic index.
administration of IGF-1 or administration of Gly-Pro-Glu, an N-terminal (c) To assess the efficacy of proposed compound in reducing angio­
peptide of IGF-1, attenuated loss of TH-immunoreactive cells in response genesis, capillary tube formation assay was done. Briefly, 120 µl
to 6-OHDA infusion into the dopaminergic axons within the nigrostriatal of diluted (10 mg/ml) growth factor reduced Matrigel was tiled
pathway[20]. Interestingly, insulin has also been shown to affect brain on a pre-cooled 48-well plate and incubated at 37⁰C for at least 3
monoamine metabolism and dopamine release [21]. In a study on h for polymerization. 35,000 HUVECs/well were seeded on so­
dopamine beta hydroxylase knockout (Dbh-/-) mice IGF-1 receptor lidified Matrigel with L-Dopa only, IGF-1 only and L-Dopa+IGF-1
phosphorylation significantly decreased as compared to their hetero­ in combination. Each group was taken in triplicate. After 16 h,
zygote littermates [22] and combination of IGF-I and bFGF reduced the tube formation was observed, and images were captured.
death rate of dopaminergic neurons from programmed cell death in
another study [23]. Thus, this suggests that even if IGF-1 promotes 2.3. Chick Chorioallantoic Membrane (CAM) assay
retinal neurogenesis, there is enough reason to believe that it can also be
neuroprotective in nature. The fertilized eggs were obtained from Kegg Farms Private Limited,
While current procedures such as laser surgery or anti-angiogenic Gurgaon and incubated at 37 ◦ C for eight days and divided into groups of
injections of VEGF antibodies are often used to arrest neo­ control (saline only), hyperglycemia (50 mM), L-Dopa alone (10 µM),
vascularization, we suggest a non-invasive approach that would IGF-1 (100 nM) alone and L-Dopa+IGF-1 (10 µM + 100 nM) (two eggs/
modulate and find a balance between neuromodulatory growth factors group). Briefly, a small window was made in the shell on day 9 of chick
and those responsible for proangiogenic activities in PDR [24]. So far, embryo development, in a laminar hood under aseptic conditions. The
different approaches have been used to prevent and/or delay neo­ window was resealed with adhesive tape after administering the drug
vascularization but simultaneous regulation of dopamine and IGF-1 treatments (100 µl/egg) and eggs were returned to the incubator until
levels has never been thought of as a therapeutic strategy. The above chick embryos were harvested on day 12. The experiment was repeated
cited literature on the influence of IGF-1 on dopamine dependent twice. Imaging was done on the same day, and eggs were subsequently
behavior of the neuronal cells in brain or in-vitro, unravels a new discarded.
approach to treat diabetic retinopathy related dopamine hypofunction
in retina. Also, since causal mechanisms intrinsic to 2.4. Animal maintenance
hyperglycemia-induced microvascular events in retina result in
increased severity of this disease, we aim to present a directed strategy Male wistar rats weighing around 120–140 g were kept under stan­
targeting neovascularization of retinal vessels and its underlying dard conditions with food and water available ad libitum. Our protocols
mechanism to slow down PDR by, restoring the activity of dopaminergic were in accordance with ARVO Statement for the Use of Animals in
neurons through administration of L-Dopa in combination with IGF-1. Ophthalmic and Vision Research and duly approved by the CPCSEA
designated Institutional Animal Ethics Committee at Amity University,
2. Methods Noida, India (Approval no. CPCSEA/IAEC/AIP/017/03/27).

2.1. Cell culture 2.5. Dosing of animals

The human retinal pigment epithelium cells (ARPE-19) were a gift A total of 80 rats were used, divided into groups of 50 and 30 as two
from Dr Anil Tiwari, Shroff Eye Hospital, New Delhi. The cells were sets, for experiments conducted at two separate time intervals and the
grown in a medium comprised of a 1:1 ratio of Ham’s F12 medium and experiments were repeated twice. The first lot of 50 animals used for
Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% morphological study, was equally divided into 4 groups namely control,
fetal bovine serum (Gibco, USA) and 1% penicillinstreptomycin com­ diabetes mellitus (DM)-treated, DM-L-Dopa-treated and DM-L-Dop­
bination (Himedia, India) according to the supplier’s instructions. a+IGF-1-treated, each containing 12 animals. Control group received
Human umbilical vein endothelial cells (HUVEC) were a gift from Dr citrate buffer (vehicle only). Other three groups (except control)
Subrajit Biswas, Amity Institute of Molecular Medicine and Stem Cell received streptozotocin (STZ; 40 mg/kg; SRL India) made in 0.1 M cit­
Research,Noida. The cells were cultured in MCDB 131 medium sup­ rate buffer, intraperitoneally twice, in a gap of two weeks [26].
plemented with 50 μg/ml endothelial cell growth supplement, 20% fetal Monitoring of blood glucose (BG) levels using a glucometer (Dr
bovine serum,2 mM L-glutamine, 50 μg/ml heparin and antibiotics Morepen) during a fasting state, 2 days after injection was done. Animals
(10,000 U/ml Pen-strep). Cell cultures were maintained at 37⁰C in a with BG level ≤ 250–300 mg/dL were considered diabetic and main­
humidified chamber with 5% CO2 in a CO2 incubator. All experiments tained. Weight and BG level of all rats was recorded twice weekly.
using HUVECs were conducted between 2 and 5 passages. At the time of Experimental term was set at 12 and 16 weeks and animals from all 4
experiments, MCDB131 medium with 2% FBS was used. groups were sacrificed at termination of each time point. L-Dopa treat­
ment began two weeks after diabetes onset and lasted until 16 weeks
2.2. Cell Cytotoxicity and in-vitro analysis post hyperglycemia. Rats from L-Dopa alone and L-Dopa+IGF-1 groups
were given i.p injections of L-Dopa (10 mg/kg; Sigma-Aldrich Corp.)
(a) The ARPE-19 cells were seeded at a density of 10,000 cells/well prepared fresh daily, in 0.1% ascorbic acid made in saline. All injections
on a 96-well plate in complete media and incubated overnight at were given 4–8 h after light onset. Rats of DM-L-Dopa+IGF-1-treated
37 ◦ C, 5% CO2. The following day, cells were pre-treated with L- group received intravitreal dose of IGF-1(100μg) (2 µl/eye; Sigma-

2
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Aldrich Corp.) twice each, after a gap of 7 days in the penultimate weeks 10 µl volume with PowerUp SYBR green 2 × Master Mix (Applied Bio­
before their sacrifice. 2 animals (from n = 50) that could not survive systems) in a StepOne RT PCR system. Each experiment was repeated
prolonged periods of diabetes were excluded from the study (Supple­ thrice. Primers were designed using Primer-BLAST NCBI (Table 1). Large
mentary Fig. 1). ribosomal protein P0 (RPLP0) and GAPDH were used as reference genes.
The parameters were set as follows: 50 ◦ C for 2 min, 95 ◦ C for 2 min, and
2.6. Hematoxylin and Eosin staining 40 amplification cycles of 95 ◦ C for 15 s and 60 ◦ C for 30 s, 95 ◦ C for 15 s,
60 ◦ C for 1 min and 95 ◦ C for 15 s for melt curve. For each cDNA sample-
Rats were euthanized by cervical dislocation. A pair of eyeballs from primer pair combination, two replicates were performed, and average Ct
each group (both time points) were fixed in 10% formalin. After 2 h, calculated. Expression of each gene of interest was calculated from the
eyecup was formalin fixed for 4 h following a protocol with slight average PCR cycle thresholds using the 2− ΔCt method.
modifications [27]. 5 µm thick sections were cut using a cryomicrotome
(Leica CM 1520) parallel to edge of the eyeball through optic disc taken 2.10. Western Blot assay
from four eyeballs. Sections were stained with hematoxylin and eosin
and viewed under microscope. Ten stained sections, lying in middle Four eyes/group (both time points) were used for protein estimation.
when plotted from optic disc to outer periphery of retina, (from each The total proteins were extracted from the retina of rats using RIPA lysis
eyeball) were selected for image acquisition at 20X. Using image anal­ buffer at 4 ◦ C. The concentration of tissue protein was determined by
ysis software Fiji, total thickness of IR was taken as thickness of ganglion Bradford assay. Sodium dodecyl sulfate-polyacrylamide gel at concen­
cell layer (GCL)+thickness of inner plexiform layer (IPL). Using 2 sec­ trations of 10% was used to separate the tissue protein by gel electro­
tions per group and examining 3 sides from each section, for three phoresis, and polyvinyl difluoride membrane-containing proteins were
randomly selected fields of view, from each section were calibrated at blocked with milk solution (5%) and/or bovine serum albumin for 60
40X magnification. All observations were made by a masked min. The cellular membrane was incubated with primary antibodies, Akt
investigator. (1:1000;Cat#4060, Cell signaling Technology, USA), phospho Akt
(1:1000;Cat#9271,Cell signaling Technology, USA),ERK(1:2000;
2.7. Transmission electron microscopy Cat#9102, Cell signaling Technology, USA), phospho ERK(1:1000;
Cat#3510, Cell signaling Technology, USA),and β-actin (1:1000;Cat#
Two eyeballs each, from both time points were analyzed (without PA5–85271,Invitrogen,USA) overnight at 4 ◦ C. Membranes were incu­
repetition). Anterior segments were removed and fixed in pre-cooled bated with peroxidase-conjugated secondary antibodies, and Image
2.5% glutaraldehyde, followed by 1% osmium tetroxide. Samples were Quant LAS 500 was used to capture the images. Fiji (Image J) was used
dehydrated using graded ethanol and embedded (Araldite CY 212). to determine mean fluorescence intensity of bands.
Ultra-thin sections (80 nm) were contrasted with uranyl acetate and lead
citrate and viewed under a Tecnai G2–20S twin transmission electron 2.11. Immunohistochemistry
microscope.
Two eyes per group were used for immunolabelling and processed
2.8. Sample collection and Dopamine analysis using a modified protocol [24]. Sections were washed and blocked in
10% normal goat serum (NGS) for 1 h. Incubation in primary antibody,
Set of 30 animals were used for serum and tissue dopamine analysis. VEGFR1 (PA5–16487,1:200, Invitrogen) prepared in 0.01 M PBS con­
Six eyes per group each were used at both time points. 6 animals (from n taining 10% NGS and 0.1% triton X-100 at 4 ◦ C overnight. After
= 30) that failed to become diabetic or could not survive prolonged washing, sections were incubated with secondaries, goat anti-rat Alexa
periods of diabetes were excluded from study. Fluor 568 (1:500, Invitrogen, #A11011) and DAPI (4′ ,6-Dia­
Dopamine estimation from tissue: Retinae from six eyes (3 ani­ midino-2-phenylindole) (1:500, Sigma, MO, USA) for 1 h. Imaging was
mals/ group) were pooled for dopamine estimation, with slight modi­ done using confocal microscope (Nikon Confocal A1).
fications to existing protocol [28]. Dissected retinae were homogenized
in 500 µl of ice cold perchloric acid containing 10 mM EDTA and 10 mM 2.12. Statistical Analysis
sodium metabisulfite, with 2.5 ng 3,4-dihyroxybenzylamide (DHBA) as
an internal standard and centrifuged at 14000 rpm for 15 min at 4ºC. Statistical analysis was performed using Graphpad Prism 8.2.144 and
Supernatant was filtered using a 0.45 µm membrane (Merck Millipore) Microsoft excel. Data are expressed as the mean ± SEM of percentage of
and analyzed. cell toxicity with respect to control. For dopamine analysis in serum and
Dopamine estimation from serum: Serum dopamine level was tissue, two-way ANOVA was performed to determine whether retinal
estimated according to established protocol with slight modifications dopamine content differed significantly between the experimental
[29]. Dopamine signals were quantified using a detection curve estab­ groups followed by Bonferroni’s post-hoc analysis (p < 0.05 *). For RT-
lished by standards ranging from 2 to 20 ng/ml. Injection volume of 20 PCR analysis, multiple Student’s t tests were used to determine whether
µl was injected into equilibrated column and modified mobile phase the expressions of the treatment groups were different from the control
consisted of 0.1 M sodium phosphate, 0.1 mM EDTA, 0.35 mM sodium group. Western blot and mean fluorescence intensity were analyzed by
octyl sulphate and 20% acetonitrile, with flow rate set at 1 ml/min. Student t test to compare the values obtained from two groups (two-
Tissue and serum dopamine levels were calculated by comparing peak tailed) while the comparison between three or more groups was
heights with those of standards. Dopamine content estimated was analyzed by one-way ANOVA. All analyses were performed with a priori
expressed as pg/mg of wet tissue and pg/ml of serum respectively. significance set at α < 0.05 (* p < 0.05; ** p < 0.01; *** p < 0.001).

2.9. Quantitative RT-PCR 3. Results

Two eyes/group at both time points were used for RNA extraction 3.1. ARPE-19 cell response to L-Dopa+IGF-1 treatment
using TRIzol (Thermo Fisher Scientific). For each sample, 1 μg of RNA
was reverse transcribed to cDNA using random primers and RevertAid On analyzing cells for cytotoxicity, we found that the combination of
First Strand cDNA Synthesis kit (Thermo Fisher Scientific). Resulting L-Dopa and IGF-1 did not reduce ARPE-19 cell viability and morphology
cDNA was diluted 1:10 in nuclease free water and 1 µl was used for each when compared to control group (Fig. 1). The growth and number of
quantitative real-time PCR (RT-qPCR). RT-qPCRs were performed in a cells in treatment group of L-Dopa+IGF-1 increased from the number of

3
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Table 1
List of Primers used in the experiment.
S.No. Gene Primer Sequence (5′ − 3′ )

1 VEGF receptor 1 TTCGTCCAACTTCTGGGCTCTT CTCCTCTTCCTTCTCTTTCTCCCC


2 VEGF receptor 2 TAGCACGACAGAGACTGTGAGG TGAGGTGAGAGAGATGGGTAGG
3 Dopamine receptor D1 CGCCCAGTCTGAAAGTTCCT TGAAGAAAGGGAGCCAGCAG
4 Dopamine receptor D2 GCCAACCTGAAGACACCACT ATCCATTCTCCGCCTGTTCAC
5 Dopamine receptor D4 GAGTGCAGAAATTCAAGCCGT GGATTTGCAGTCAGGGGCTA
6 IGF-1 receptor CCAACAAGTTCGTCCACAG AGTCCGTCTCGTAGATGTC
7 Ribosomal Protein Lateral Stalk Subunit P0 AGTACCTGCTCAGAACAC TCGCTCAGGATTTCAATGG

Fig. 1. Representative images of effect of L-Dopa+IGF-1 on cell morphology after 24 h (A). (a) ARPE-19 cells were cultured in serum-supplemented basal media,
without high glucose (50 mM) (control) treatment, (b) ARPE-19 cells were cultured in serum-supplemented basal with high glucose (50 mM) and (c) ARPE-19 cells
were cultured in serum-supplemented basal with high glucose (50 mM) and combination of L-Dopa+IGF-1. (B) The dose response curves for determination of IC50
values of L-dopa and IGF-1 showed that the IC50 of L-Dopa was 69.1 μM and IGF-1 was 1.14 nM respectively.(C)The CI values of L-Dopa and IGF − 1 was found to be
less than 1, which shows synergistic effect of the drug. Scale bars denote 500 µm at 40 × magnification.

cells seen in HG-treated group. 3.3. Reduced angiogenesis obtained in the combination group when
The pre-treatment of L-Dopa alone and IGF-1 alone after hypergly­ assessed in vitro by CAM assay
cemia, resulted in reduced number of cells with increasing concentra­
tions when compared to control (not shown here). The IC50 of L-Dopa Chick chorioallantoic membrane (CAM) assay is one of the most
was found to be 69.1 µM and that of IGF-1 was 1.41 nM. For estimation reliable assays known for studying angiogenesis (Fig. 3). Visualized for
of optimal combination ratio for maximal synergy, the IC50 of L-Dop­ its effects after 48 h, the analysis of blood vessels in CAM was done based
a+IGF-1 was tested in two combinations: combination I (IGF-1 on thickness of vessel, branching, and sprouting of vessel. We found that
100 nM+L-Dopa 10 μM) and combination II (IGF-1 100 nM+L-Dopa the combination of L-Dopa+IGF-1 considerably reduced angiogenesis
30 nM) respectively. The combination index study revealed that the more than the drugs used alone.
combination of L-Dopa and IGF-1 acted synergistically at IC50 values for
combination I (IGF-1 100 nM+L-Dopa 10 μM). The CI value was 0.57
which is less than 1 and indicates that the two drugs in combination had 3.4. Intravitreal IGF-1 improves microstructural differences and
a synergistic effect on cells. morphometry in diabetic retina

3.2. Effect of L-Dopa+IGF-1 on viability and tube formation of HUVECs The changes in blood glucose levels (Supplementary Table 1) and
weight of rats at both time points were duly recorded (Supplementary
To investigate the effect of L-Dopa+IGF-1 on high-glucose-induced Table 2). Morphological changes when compared with respective age
endothelial cells injury, we treated HUVECs with combination of L- matched controls (Fig. 4) showed a decrease in number of RGCs in inner
Dopa+IGF-1 for 24 h after 24 h high glucose treatment (50 mM) nuclear layer (INL) and outer nuclear layer (ONL), with increased
(Fig. 2). MTT assay showed that L-Dopa+IGF-1 partially prevented high- vascular permeability and loss in structural integrity, resulting in RGC
glucose-induced endothelial cells injury better than IGF-1 alone and necrosis initiation in DM-treated group of 12 weeks (Fig. 4A). RGCs with
comparable to L-Dopa alone (Fig. 2a). shrunken pyknotic nuclei and surrounded by new vessels appeared in
junctions between GCL and IPL and between IPL and INL in DM-treated
group with visible basal laminar thickening in 16 weeks (Fig. 4B). In

4
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Fig. 2. Representative image of effect of L-Dopa supplemented with IGF-1 on morphology (A), tube formation capability (B) and viability of human umbilical vein
endothelial cells (HUVECs) (C). (A) Cells showing normal morphology in control group supplemented with vehicle(a). Cells treated with HG at 50 mM for 24 h (b),
cells treated with HG at 50 mM and IGF-1 (c) and cells treated with L-Dopa+IGF-1 after HG (d). (B) Cells in control with no treatment (a), Cells treated with HG alone
(b), HG-IGF-1 alone (c) and HG-L-Dopa+IGF-1 (d) for tube formation assay. (C) Cells in control with no treatment, cells treated with HG alone, HG-IGF-1 alone and
HG-L-Dopa+IGF-1 for 24 h, the cell viability was determined by MTT assay. These results revealed that L-Dopa alone and L-Dopa supplemented with IGF-1 increased
cell viability of HUVECs significantly (p < 0.05 *). Each bar represents mean±SEM where n = 3. *p < 0.05 vs control and DM-treated. Scale bar equals 500 µm.

Fig. 3. Representative photographs showing


the anti-angiogenic effect of different treatment
groups on chick embryo chorioallantoic mem­
brane (CAM). (a) Representative image for
positive control (saline). (b) Representative
image for negative control (High glucose (HG)
at 50 mM). (c) Representative illustration of the
inhibition of angiogenesis in HG-treated-L-Dopa
where angiogenesis reduced compared to HG
treated group. (d) IGF-1 treated group showed
more vessels compared to HG-treated-L-Dopa
alone treated group. (e) Significant reduction in
number of vessels was seen in HG treated-L-
Dopa+IGF-1-treated group.

5
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Fig. 4. Representative photomicrographs of hematoxylin and eosin-stained retinal sections observed at 12 weeks (A) and 16 weeks (B). (A) In 12 weeks rats (a)
Normal retinal layers in control group. (b) DM-treated group showing decreased RGCs, increased diameter of retinal vessels and presence of micro vessels with visible
thinning of IR. (c) DM-L-Dopa-treated group showing ballooning degeneration with pyknotic nuclei and mild edema. (d) DM-L-Dopa+IGF-1-treated group showing
better number of RGCs with restored thickness of IR. (B) In 16 weeks, control group with linear arrangement of layers (a) and DM-treated group showing distorted
morphology of GCL with vacuolated ganglion cells and decreased thickness of IR (b) was visible. DM-L-Dopa-treated group showing improved arrangement of GCL
with some micro vessels (c) and (d) DM-L-Dopa+IGF-1-treated group showing restored IR thickness and less or no presence of micro vessels was observed. (C)
Quantification of IR thickness measured on retinal sections. The graph shows variations in thickness of IR (GCL+IPL) scanned over a period of 16 weeks. N = 4.Each
bar represents mean±SEM. p * ** *< 0.05, * **p < 0.05 and * *p < 0.05 vs DM-treated (negative control).

Fig. 5. Representative photomicrographs of ultrastructure of retinal capillary at 12 weeks (a-d) and 16 weeks (e-f) respectively. Retinal vessels were observed to be
normal in IPL and INL of 12 weeks control group (a); STZ only treated DM group (b); diabetic rats subjected to L-Dopa alone (c); diabetic rats subjected to com­
bination of L-Dopa+IGF-1 (d) retinal capillary of control group at 16 weeks (e); deformed RBCs in DM-treated group (f); DM-L-Dopa-treated (g) and realignment of
BM as seen in DM-L-Dopa+IGF-1-treated groups (h). Scale bar equals 1–5 µm. H=heterochromatin; Ed=endothelial cells; BM=basement membrane; RBC=red blood
cell; SM=smooth muscle; black arrow=capillary lumen; P = pericyte.

6
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

treatment groups of DM-L-Dopa-treated and DM-L-Dopa+IGF-1-treated, 3.6. Dopamine level changes significantly between DM-treated and DM-
few scattered blood vessels and maintained continuity of GCL with DA+IGF-1 treated conditions
reduced loss of RGCs was found in both 12 and 16 weeks.
Thinning of IPL, INL and ONL occurs due to desquamation of cells High performance lipid chromatography analysis of samples signifies
with multiple pyknotic nuclei and widening of intercellular spaces in changes in dopamine content of control, DM-treated, DM-L-Dopa-
RGCs of DM group (Fig. 4C). Damage caused by loss of RGCs and scat­ treated and DM-L-Dopa+IGF-1-treated groups at both 12 weeks and 16
tered endothelial cells became more prominent in 12 weeks, but treat­ weeks in retinal tissue and serum. Dopamine level of control rats
ment modality proved effective with significant difference between DM- sacrificed at 12 weeks was 111 ± 1 ng/mg of wet tissue (Fig. 6A) but
treated and DM-L-Dopa+IGF-1-treated groups’ IR thickness (80 ± 1.8 vs went down to 34 ± 0.8 ng/mg of wet tissue in DM-treated group. On
106 ± 2.7, ****p < 0.05). Meanwhile, new vessels in junction between treatment with L-Dopa alone, the levels in DM-L-Dopa-treated retina
GCL and IPL and those between IPL and INL in retinae of 16 weeks, were restored to 59 ± 0.4 ng/mg of wet tissue (*p < 0.05). Animals
resulted in significant variation in IR thickness (66.2 ± 5.6 vs 95.9 receiving IGF-1 with L-Dopa showed increased dopamine level at 54
± 8.6, **p < 0.05) between DM-treated vs DM-L-Dopa+IGF-1-treated ± 0.4 ng/mg of wet tissue (*p < 0.05). In comparison, 16 weeks dopa­
retinas respectively. mine levels varied from 30 ± 0.33 in control to 3.8 ± 0.2, 23.2 ± 0.3
RGC, retinal ganglion cells; GCL, ganglion cell layer; IPL, inner (*p < 0.05) and 25.3 ± 0.5 (*p < 0.05) in DM-treated, DM-L-Dopa-
plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; treated and DM-L-Dopa+IGF-1-treated groups, respectively.
ONL, outer nuclear layer. Serum levels of dopamine were also checked to ensure that they
corroborate with results of tissue specimen. Dopamine content in con­
3.5. Ultrastructure changes trol, DM-treated, DM-L-Dopa-treated and DM-L-Dopa+IGF-1-treated
groups was found to be 9.7 ± 0.1, 7.5 ± 0.14, 8.7 ± 0.2 (*p < 0.05) and
Pair of eyeballs from each group was studied to see how L-Dopa with 8.3 ± 0.03 pg/ml of serum at 12 weeks (Fig. 6B). At 16 weeks, dopamine
IGF-1 affects ultrastructure of capillaries in the transition period of the content in control, DM-treated, DM-L-Dopa-treated and DM-L-Dop­
two time points (Fig. 5). In 12 weeks, control, BM was complete with a+IGF-1-treated group was found to be 5.2 ± 0.001, 1.5 ± 0.1, 3.15
unobstructed lumen (Fig. 5a). In DM-treated group, stratified BM, ± 0.01(*p < 0.05) and 4.38 ± 0.12 (*p < 0.05) ng/ml of serum
bulging lumen with distorted smooth muscle was observed (Fig. 5b). respectively. A reduction in dopamine levels of control groups of both
Whereas, in DM-L-Dopa-treated animals, lumen gradually smoothened tissue and serum in 16 weeks was seen as basal dopamine level is known
with progressive improvement in BM (Fig. 5c) and optimal protective to reduce in rats with aging [30].
effects of the treatment were visible in DM-L-Dopa+IGF-1-treated group
where smoothened BM, with some displaced pericytes were seen
(Fig. 5d). 3.7. Variations in expression of VEGF receptor 1, VEGF receptor 2 are
Retinal capillary BM, endothelial cells, and pericytes of rats at 16 modulated by changes in IGF-1 receptor levels and dopamine receptor
weeks had normal morphological structures without apparent patho­ levels
logical changes in control group (Fig. 5e). Thickened and discontinuous
BM was observed. Endothelial cells turned hyperplasic and bulged into Variation in mRNA expression of VEGF receptor 1 and VEGF receptor
lumen and pericytes became confined to capillary wall or were lost 2 in rat retinas were analyzed after diabetes initiation. We found the
(Fig. 5f). Furthermore, capillary BM of treatment groups of DM-L-Dopa- expression of VEGF receptor 1 was increased in rat retina at 12 weeks
treated and DM-L-Dopa+IGF-1-treated were complete and continuous [27,31] but became weak at 16 weeks after development of diabetes
while degree of thickening also improved. Endothelial cell proliferation (p < 0.05 *). Increased expression of VEGF receptor 1 was attenuated by
decreased, lumen bulging reduced and pericytes showed sporadic edema both DM-L-Dopa-treated and DM-L-Dopa+IGF-1-treated groups
(Fig. 5g), with significantly improved capillary seen in DM-L-Dopa+IGF- (*p < 0.05) at 12 weeks but significantly only in DM-L-Dopa-treated
1 treated group (Fig. 5h). group in 16 weeks (*p < 0.05) (Fig. 7A). Expression levels of VEGF re­
ceptor 2 after initial increase in DM-treated groups of both 12 and 16
weeks [32] reduced significantly in both DM-L-Dopa-treated and
DM-L-Dopa+IGF-1-treated groups of both time points (Fig. 7B).
Dopamine D1 receptor, D2 receptor and D4 receptors were used to
assess changes in their respective transcript levels to further confirm

Fig. 6. Effect of STZ on concentrations of dopamine in (A) retina (pg/mg wet tissue) and in (B) serum (pg/ml) at 12 and 16 weeks after diabetes and treatment with
L-Dopa and L-Dopa+IGF-1. Each bar represents mean±SEM where n = 3. *p < 0.05 vs DM-treated.

7
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Fig. 7. Quantitative real-time RT-PCR analysis of (A)VEGF receptor 1, (B)VEGFR receptor 2, (C) dopamine D1 receptor, (D) dopamine D4 receptor, (E) dopamine D2
receptor and (F) IGF-1 receptor from DM-treated, DM-L-Dopa-treated and DM-L-Dopa+IGF-1-treated groups (n = 4, each group) and normal controls (n = 4). The
mRNA levels were normalized to RPLP0 housekeeping gene. Each bar represents mean±SEM, *p < 0.05.

counteracting effect of L-Dopa+IGF-1 treatment on diabetes (Fig. 7C). 3.8. L-Dopa+IGF-1 alleviates streptozotocin induced degeneration
The expression of dopamine D1 receptor increased in DM-treated groups
of 12 as well as 16 weeks but decreased significantly (*p < 0.05) in DM- To understand the molecular mechanisms involved in overcoming
L-Dopa-treated and significantly in DM-L-Dopa+IGF-1-treated group. At the effect of stz damage on retina, we examined both ERK and Akt
the same time expression of dopamine D4 receptor increased in DM pathways, since they are the key signaling pathways regulating cell
group of 12 as well as 16 weeks which then reduced in both DM-L-Dopa- growth and survival but aberrantly activated in diseases like cancers and
treated and DM-L-Dopa+IGF-1-treated groups but significantly diabetic retinopathy (Figs. 8 and 9). Western blot analysis of ERK and
(*p < 0.05) only in DM-L-Dopa-treated group. Contrary to this, the phospho-ERK (Fig. 8A) revealed, by densitometric analysis of four
expression of dopamine D2 receptor decreased in 12 as well as 16 weeks paired samples in each experimental group (given in parentheses as the
of DR in DM-treated group. Expression of dopamine D2 receptor mean ± SE), that L-Dopa without treatment resulted in slightly
increased significantly in DM-L-Dopa+ IGF-1-treated group of 12 weeks increased level of ERK (Fig. 8B) and significant increase in expression of
with increase also seen in 16 weeks in both L-Dopa-treated groups but p-ERK (Fig. 8C) in DM-treated groups. The combination of L-Dopa+IGF-
significantly (*p < 0.05) only in DM-L-Dopa-treated group of 16 weeks 1 reduced ERK and p-ERK expression considerably when compared with
(Fig. 7D). control and DM groups. This suggests that ERK pathway mediates
The expression of IGF-1 receptor decreased in diabetic rats at 12 as changes in diabetic retina after STZ induced damage and is overcome
well as 16 weeks in DM-treated group. Increase in the expression of IGF- considerably by treatment of L-Dopa+IGF-1.
1 receptor in the DM-L-Dopa-treated group in 12 weeks but decreased Analysis of protein levels of Akt and p-Akt (Fig. 9) revealed that Akt
significantly in 16 weeks (Fig. 7E). On application of IGF-1 along with L- (Fig. 9B) and p-Akt levels increased in DM-treated group as compared to
Dopa, IGF-1 receptor levels did not change significantly in 12 weeks but control (Fig. 9C). In the L-Dopa alone group, expression of Akt increased
increased significantly in 16 weeks (Fig. 7F). and p-Akt decreased compared to control. L-Dopa+IGF-1 decreased both
Akt and p-Akt expression significantly when compared to control and

8
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Fig. 8. Western blotting determined the expression of ERK and p-ERK following treatment with L-Dopa alone and L-Dopa+IGF-1 after diabetes induction in rats of 16
weeks (A). The quantitation of ERK (B) and p-ERK (C) was determined by normalizing to β actin. Data are presented as mean±SE of mean from two different ex­
periments.*p < 0.05 .

Fig. 9. Western blotting determined the expression of Akt and p-Akt following treatment with L-Dopa alone and L-Dopa+IGF-1 after diabetes induction in rats of 16
weeks (A). The quantitation of Akt (B) and p-Akt (C) was determined by normalizing to β actin. Data are presented as mean±standard error of mean from two
different experiments.*p < 0.05 .

9
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

Fig. 10. Immunohistochemistry of retinal sections showing VEGF receptor 1 expression (A) and mean fluorescence intensity (B). (A) control (a-c), DM (d-f), and
treatment group of DM-L-Dopa+IGF-1 receptor (g-i) respectively. Bars represent mean ± SEM of at least 2 independent experiments. Scale bar= 100 µm.*p < 0.05 .

also comparable to L-Dopa alone. diabetic retinopathy, to further establish the protective function of
L-Dopa when combined with IGF-1 which, is also speculated to possess
neuroprotective function [38]. We found a significant positive correla­
3.9. Immunofluorescence studies suggest that VEGF receptor 1 expression
tion between progression of diabetic retinopathy and decreased dopa­
lowers with L-Dopa+IGF-1 combination
mine levels. When L-Dopa was supplemented with IGF-1, it acts in
tandem with L-Dopa to overcome neurodegenerative and vascular
Previously it has been shown that expression of VEGF receptor 1
changes, often encountered in late-stage retinopathy.
increased in retinal vasculatures of diabetic rats [33] and blockade of
In order to check whether the dose of L-Dopa and IGF-1 to be used in
VEGF receptor 1 reduces inflammation thereby preventing leakage [31],
animals does not prove to be cytotoxic, we evaluated cell viability by
but it remains unknown as to how its expression is affected by IGF-1
MTT assay on ARPE-19 cells. The proliferation of ARPE-19 cells was
stimulation with L-Dopa in diabetic rats. We therefore examined VEGF
inhibited by high glucose at 50 mM. L-Dopa+IGF-1 at a concentration
receptor 1 expression in diabetic rat retina (Fig. 10) and found that
range of 0–50 µM, did not endanger the survival and morphology of
VEGF receptor 1 expression was increased in DM-treated groups of 16
ARPE-19 cells (Fig. 1). Even though the anti-angiogenic potential of L-
weeks (12 weeks not shown here) (Fig. 10d-f) compared with nondia­
Dopa has already been established in several studies [39,40], the ability
betic controls where there was no or barely detectable immunoreactivity
of L-Dopa to ameliorate proangiogenic activity of IGF-1 in proliferative
(Fig. 10a-c) and immunofluorescence staining further confirmed its
diabetic retinopathy has not been explored yet. In this regard, when
vascular localization. Increased vascular effect was noted to have
HUVECs were treated with L-Dopa+IGF-1, the cell morphology did not
decreased in DM-L-Dopa+IGF-1-treated group (Fig. 10g-i).
differ much from those treated with IGF-1 alone, (Fig. 2 A) and the cell
viability showed significant improvement when compared to the DM
4. Discussion
group (p < 0.05 *) (Fig. 2C). As the endothelial cell tube formation
assay is a useful method to determine genes and pathways that poten­
As the dopaminergic system is a promising target for developing
tially play an important role in activation or inhibition of angiogenesis
strategy against diabetic retinopathy [34], dopamine precursor L-Dopa,
[41], we examined the potential of our drug treatment of L-Dopa+IGF-1
has been found to be neuroprotective against early retinal dysfunction
against IGF-1 alone in HUVEC cells. The density of the network of tu­
associated with diabetic retinopathy in both diabetic humans and ani­
bules formed by endothelial cells was very robust in DM group or hy­
mals [5,17,35,36] and also on neovascularization in cancer [37]. We
perglycemic condition. When exposed to DM-L-Dopa+IGF-1, the
wanted to analyze the efficacy of L-Dopa with IGF-1 in late stages of

10
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

network began to loosen and break as compared to that seen in IGF-1 Multiple studies have concluded that accelerated retinopathy results
alone (Fig. 2B). from increased IGF-I production and this leads to development of PDR
Since, the CAM is a highly vascularised membrane found in fertilized [50,56,57]. In recent studies IGF-1 mRNA was found to be reduced in
chicken eggs, with a vast vascular network of capillaries, veins and ar­ eye in early stages [6,58,59] and it has been suggested that exogenous
teries, it can be easily manipulated and observed for experimental study treatment with IGF-1 may be beneficial [58]. Since it has been proven
of angiogenesis [42]. The use of the CAM assay as an efficient biological that IGF-1 is antiapoptotic and enhances nerve metabolism [60], we
screening tool on the focus of cell induced angiogenesis is important and found that L-Dopa and IGF-1 co-administered in combination could
hence, when we tested our combination of L-Dopa+IGF-1 against downregulate VEGF receptor 1/VEGF receptor 2 activation witnessed in
L-Dopa alone or IGF-1 alone, we found that the number of vessels 16 weeks DM-treated group (Fig. 7 A and B). Since studies regarding
reduced considerably from those seen in positive control (Fig. 3). modulation of synthesis of IGF-1 or blocking IGF-1 could attenuate
Adequate dopamine levels result in normalization of morphology retinal angiogenesis [61] we also found that IGF-1 when combined with
and improved vessel function by acting on pericytes and endothelial L-Dopa might reduce ability of IGF-1 to induce VEGF receptor expres­
cells [36] and when we observed the treatment groups of DM-L-Dopa-­ sion. Some reports where mRNA and protein expression of VEGF re­
treated and DM-L-Dopa+IGF-1-treated for morphological differences, ceptor 1 and receptor 2 was seen in normal retina [61–63] suggest
we found improved structural features which can be attributed to physiological functions of VEGF, but few data is available on expression
dopamine biosynthesis aided by L-Dopa(Fig. 4). We chose data points to patterns of VEGF receptors in the retinal vasculature of control or
be a maximum of 16 weeks to better assess contrasting opinions on DM-treated eyes [35,44,55,62].
characterization of early and late-stage diabetic retinopathy as, symp­ The expression of IGF-1 receptor and VEGF receptors is interrelated
toms of neovascularization like acellular capillaries, ghost like pericytes with respect to the prevalence of retinopathy and occurrence of neo­
occurs between 16 and 36 weeks, and vascular changes are visible as vascularization. In postnatal stages during artificially induced retinop­
early as 2 weeks of hyperglycemia [43,44]. Since, neuronal defects are athy, IGF-1 mRNA levels are low in humans, mice, and diabetic rats after
characterized by reduction in thickness of inner retina [45], we observed 5 months of diabetes and show that IGF-1 levels are much less than those
that at 12 weeks, marked morphological alterations like reduced in control [64]. Although activation of IGF-1R initiates cascade of PI3/
thickness and breakage in all inner layers were visible (Fig. 4A). A pAkt pathway and expression of phospho-VEGF [65] but increase in
definite increase in vascular lesions due to increased vessel permeability IGF-1 mRNA levels in retinopathy condition is also associated with
were visible at 16 weeks of diabetes (Fig. 4B). Consistent with previous suppressed VEGF mRNA levels [66]. The expression of IGF-1 receptor
studies where decreased IR thickness is characterized as primary pa­ was much less than control in both 12 week and 16-week time points in
thology in diabetic retinopathy, much before appearance of vascular DM-treated group (Fig. 7F) and levels of VEGF receptor 1 and VEGF
lesions [46], significant decrease in thickness of IR was overcome by receptor 2 increased in DM-treated group as described previously [21].
treatment with L-Dopa+IGF-1 better than L-Dopa alone (Fig. 4C) when In addition, increased VEGF mRNA levels in ischemic retina of patients
quantitative measurement of GCL+IPL thickness was used [47–49]. with advanced diabetic retinopathy and proliferative diabetic retinop­
Ultrastructural analysis of retinal capillaries on treatment with L- athy is associated with high levels of VEGF in the vitreous [1,28,67]. On
Dopa or L-Dopa+IGF-1 revealed considerable recovery of capillaries administering L-Dopa, IGF-1 receptor level increased in 12 weeks due to
with intact endothelial cells, reduced microaneurysm formation, vessels increase in IGF-1 which is validated by decrease in VEGF receptor 1 and
with less deformed RBCs and reduced BM thickening in both 12 and 16 VEGF receptor 2 expressions at 12 weeks (Fig. 7A and B). At 16 weeks of
weeks (Fig. 5c, d,g and h) which could only be analyzed but not quan­ diabetes when neovascularization started, IGF-1 receptor levels were
tified here. Such changes in BM and vasculature are often attributed to sufficiently low as evident from comparably higher levels of VEGF re­
protein kinase C activation in diabetic retinopathy [50]. ceptor 1 and VEGF receptor 2 and L-Dopa treatment was unable to
It has been found that L-Dopa if given at a lower dose or intermit­ enhance it. When IGF-1 was given along with L-Dopa, its level increased,
tently, may reverse measurable early-stage diabetic retinopathy [36]. although no significant change was observed for IGF-1 receptor
Since retinal dopamine reduction in diabetes originates from decreased expression in 12 weeks but significant enhancement in 16 weeks caused
biosynthesis of dopamine, we believe that exogenous supplementation downregulation of VEGF receptor 1 and to some extent VEGF receptor 2
of L-Dopa with IGF-1 further co-stimulates production of dopamine to levels.
overcome this deficiency. Level of dopamine as measured in retinal The signaling mechanisms that lead to phosphorylation of IGF-1 and
tissue and serum showed a significant decrease in animals of DM-treated in turn activate VEGF and it’s binding to VEGF receptor 1 and VEGF
group as observed previously [51] when compared to control group of receptor 2 induces activation of PI-3 K and Akt pathways [22,68]. IGF-1
both 12 and 16 weeks (Fig. 6A and B). Hence, IGF-1 supplicated with plays an important role in cell proliferation by governing PI3K/Akt
L-Dopa restored dopamine levels in both tissue and serum, in signaling and effects of which maybe mediated through the
DM-L-dopa-treated and in DM-L-Dopa+IGF-1-treated groups signifi­ Mitogen-activated protein kinase kinase/extracellular regulated protein
cantly back to comparable levels of control in agreement with a similar kinases (MAPK/ERK) signaling pathway. IGF-1 triggers ERK pathway
study [52]. activation through the Ras/Raf/MEK cascade. Activated ERK (p-ERK)
When the transcript levels of dopamine receptors D1, D4 and D2 directly activates various downstream kinases and transcription factors
were checked, increase in dopamine D1receptor and D4 receptor and these signaling pathways have been implicated in the stimulation of
expression indicates the binding of L-Dopa to these receptors is not al­ cell proliferation. We observed significant downregulation of ERK in
ways influenced by its depletion in diabetic retinopathy conditions [9, treatment group of L-Dopa+IGF-1 which, implies that the combination
53]. But application of L-Dopa with IGF-1 could restore the expression of restricts formation of new blood vessels thereby restricting progression
dopamine D4 receptor significantly to the level of control in both 12- of PDR (Fig. 8). Since, the suppression of Akt/ERK phosphorylation in­
and 16-week stages better than L-Dopa alone (Fig. 7C, D, and E). The hibits angiogenesis [69], the activation of IGF-1 leads to overexpression
expression of dopamine D2 receptor is affected by loss of dopamine in of its downstream target Akt and phosphorylates Akt. The transcript
retinopathy conditions as indicated in previous studies [54,55]. level of p-Akt in DM group could be downregulated in DM-L-Dopa and
Administration of L-Dopa+IGF-1 could restore dopamine D2 expression DM-L-Dopa+IGF-1 group (Fig. 9). It has been shown that inhibition of
levels comparable to control in 12 as well as 16 weeks and with L-Dopa IGF-1 inhibits phosphorylation of IGF-1, Akt and ERK1/2 [61]. Down­
alone in 12 weeks. This observation leads to the conclusion that regulation in Akt/p-Akt in L-Dopa or L-Dopa+IGF-1 treatment supports
downstream targets of IGF-1 in its signaling cascade at some point the concept that suppression of protein kinase B (Akt) causes inhibition
modulate, dopaminergic neurotransmission and improve the efficacy of of angiogenesis and neovascularization [70].
dopamine in restoring retinal function in retinopathy conditions. Observation of VEGF receptor 1 expression is important and normal

11
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

expression of VEGF receptor 1 in RNFL and IPL was found in control rats Conflict of interest statement
(Fig. 10a-c). After diabetes induction, VEGF receptor 1 became densely
expressed in the GCL, IPL, INL, OPL, and ONL in diabetic rats (Fig. 10d- The authors declare that they have no known competing financial
f), in conjunction with previous studies [71–74]. Abrogation of VEGF interests or personal relationships that could have appeared to influence
expression takes place in the treatment group of DM-L-Dopa+IGF-1-­ the work reported in this paper.
treated better when compared to DM-treated group (Fig. 10g-i). This
balance in the receptor expression induced by L-Dopa+IGF-1 treatment Acknowledgement
is probably what stopped/slowed neovascularization in 16 weeks and
delayed the progression of proliferative retinopathy which was also We would like to thank Science and Engineering Research Board
confirmed by measurement of mean fluorescence intensities (Fig. 10B). (Department of Science and Technology) for funding this study under
IGF-1 works to enhance dopamine levels in diabetic condition and acts the grant number EMR/2016/004054. The authors wish to thank Dr
coherently to maintain IGF-1 levels that can regulate VEGF receptor Tapas Chandra Nag and his staff and technicians at AIIMS EM facility,
expression. Low levels of IGF-1 may disrupt functioning of VEGF and New Delhi for providing technical support for TEM processing and im­
only rapid control of hyperglycemia with insulin results in rise in IGF-1R aging. We are also grateful to Dr Seema Sen from R.P Centre, AIIMS for
levels [75]. Return to normoglycemia with long-term insulin therapy providing access for processing hematoxylin and eosin sections. The
presumably normalizes VEGF and IGF-1 levels, resulting in an eventual authors would also like to thank Dr Subrajit Biswas from Amity Institute
reduction in retinopathy [76]. of Molecular Medicine and Stem Cell Research, Amity University, Noida
and his student Maryam for providing the cells and assistance in con­
5. Conclusion ducting experiments on HUVEC. We would also like to thank Dr Anil
Tiwari,Shroff eye Hospital, New Delhi for gifting us ARPE-19 cells.
We provide comprehensive proof of action of L-Dopa and IGF-1 Lastly, we would also like to thank Dr Ajit Verma, Amity Institute of
working synergistically on late-stage retinopathy to diminish the pro­ Microbial Technology, Amity University, Noida for allowing access to
fuse vascularization of the retina observed during PDR. Adequate the confocal facility.
availability of L-Dopa in the microenvironment of the retina could
reverse the proangiogenic effect of IGF-1 and in turn influence the level
Conflict of Interest
of VEGF receptors.
The effect of hyperglycemia and its subsequent abrogation through
The authors declare no conflict of interest.
treatment with L-Dopa+IGF-1 was not only established through
morphological and biochemical assays but also supported by changes in
Appendix A. Supporting information
transcript levels of mRNA and protein. Since, it is already known that
diabetic retinopathy related neuronal degeneration in retinal neurons
Supplementary data associated with this article can be found in the
occurs due to activation of phophatidylinositol 3-kinase (PI3)/Akt ki­
online version at doi:10.1016/j.biopha.2022.112868.
nase pathway, we found that L-Dopa+IGF-1 regulates PI3K/Akt
pathway which also modulates expression of VEGF receptor 1 and VEGF
References
receptor 2 and implies that suppression of angiogenesis takes place in 16
weeks. The hypothesis was also replicated in CAM model for substantial [1] J.J. Steinle, Sympathetic neurotransmission modulates expression of inflammatory
evidence of the potential of L-Dopa in combination with IGF-1 slowing markers in the rat retina, Exp. Eye Res. 84 (2007) 118–125, https://doi.org/
down or delaying neovascularization during progression to PDR. 10.1016/j.exer.2006.09.006.
[2] C.T. Motz, K.C. Chesler, R.S. Allen, K.L. Bales, L.M. Mees, A.J. Feola, A.Y. Maa, D.
This is the first study to report the anti-angiogenic effect of a pro- E. Olson, P.M. Thule, P.M. Iuvone, A.M. Hendrick, M.T. Pardue, Novel detection
angiogenic growth factor like IGF-1, when combined with L-Dopa, en­ and restorative levodopa treatment for preclinical diabetic retinopathy, Diabetes
hances its protective function and rescues the retina from PDR. Hence, 69 (2020) 1518–1527, https://doi.org/10.2337/db19-0869.
[3] C. Nishimura, K. Kuriyama, Alterations in the retinal dopaminergic neuronal
our study provides enough evidence to corroborate our claims of syn­
system in rats with streptozotocin-induced diabetes, J. Neurochem. 45 (1985)
ergsistic function of dopamine with IGF-1 in reducing adverse effects of 448–455, https://doi.org/10.1111/j.1471-4159.1985.tb04008.x.
PDR. [4] M.J. Gastinger, R.S.J. Singh, A.J. Barber, Loss of cholinergic and dopaminergic
amacrine cells in streptozotocin-diabetic rat and ins2 akita -diabetic mouse retinas,
Invest. Ophthalmol. Vis. Sci. 47 (2006) 3143–3150, https://doi.org/10.1167/
Funding iovs.05-1376.
[5] M.K. Kim, M.H. Aung, L. Mees, D.E. Olson, N. Pozdeyev, P. Michael, P.M. Thule, M.
This work was sponsored by DST-Science and Engineering Research T. Pardue, U. States, dopamine deficiency mediates early rod-driven inner retinal
dysfunction in diabetic mice, Invest Ophthalmol. Vis. Sci. 59 (2018) 572–581,
Board, New Delhi, India (EMR/2016/004054). https://doi.org/10.1167/iovs.17-22692.
[6] M.H. Aung, H. Park, M.K. Han, T.S. Obertone, J. Abey, F. Aseem, P.M. Thule, P.
CRediT authorship contribution statement M. Iuvone, M.T. Pardue, Dopamine deficiency contributes to early visual
dysfunction in a rodent model of type 1 diabetes, J. Neurosci. 34 (2014) 726–736,
https://doi.org/10.1523/JNEUROSCI.3483-13.2014.
Shikha Upreti: Methodology, Formal analysis, Validation, Data [7] H.W. Van Dijk, P.H.B. Kok, M. Garvin, M. Sonka, J.H. Devries, R.P.J. Michels, M.E.
curation, Investigation, Writing - review & editing. Seema Sen: Data J. Van Velthoven, R.O. Schlingemann, F.D. Verbraak, M.D. Abra, Selective loss of
inner retinal layer thickness in type 1 diabetic patients with minimal diabetic
curation, Investigation, Resources. Tapas Chandra Nag: Writing - re­ retinopathy, Invest. Ophthalmol. Vis. Sci. 50 (2009) 3404–3409, https://doi.org/
view & editing. Madhumita P. Ghosh: Conception, Design, Methodol­ 10.1167/iovs.08-3143.
ogy, Resources, Data curation, Writing - review & editing, Project [8] R. Brandies, S. Yehuda, The possible role of retinal dopaminergic system in visual
performance, Neurosci. Biobehav. Rev. 32 (2008) 611–656, https://doi.org/
administration, Funding acquisition.
10.1016/j.neubiorev.2007.09.004.
[9] C.R. Jackson, G. Ruan, F. Aseem, J. Abey, K. Gamble, G. Stanwood, R.D. Palmiter,
Author’s Contribution P.M. Iuvone, D.G. Mcmahon, Retinal dopamine mediates multiple dimensions of
light-adapted vision, J. Neurosci. 32 (2012) 9359–9368, https://doi.org/10.1523/
JNEUROSCI.0711-12.2012.
SU: Methodology, Formal analysis, Validation, Data curation, [10] I. Nir, R. Haque, P.M. Iuvone, Diurnal metabolism of dopamine in dystrophic
Investigation, And Writing - review and editing. SS: data curation, retinas of homozygous and heterozygous retinal degeneration slow (rds) mice,
investigation, and resources. TCN: writing - review & editing. MG: Brain Res 884 (2000) 13–22, https://doi.org/10.1016/s0006-8993(00)02855-9.
[11] S.E. Doyle, W.E. Mcivor, M. Menaker, Circadian rhythmicity in dopamine content
conception and design, methodology, resources, data curation, writing - of mammalian retina: role of the photoreceptors, J. Neurochem. 83 (2002)
review & editing, project administration and funding acquisition. 211–219, https://doi.org/10.1046/j.1471-4159.2002.01149.x.

12
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

[12] H. Lahouaoui, C. Coutanson, H.M. Cooper, M. Bennis, O. Dkhissi-benyahya, Krüppel-like factor-2 expression in tumor endothelial cells, Proc. Natl. Acad. Sci. U.
Diabetic retinopathy alters light-induced clock gene expression and dopamine S. A. (2011) 1–6, https://doi.org/10.1073/pnas.1108696108.
levels in the mouse retina, Mol. Vis. 22 (2016) 959–969. [38] W. Song, W. Jiang, C. Wang, J. Xie, X. Liang, Y. Sun, L. Gong, W. Liu, L. Qu,
[13] N. Inokuchi, T. Ikeda, Y. Imamura, C. Sotozono, S. Kinoshita, Y. Uchihori, Vitreous Jinmaitong, a traditional chinese compound prescription, ameliorates the
levels of insulin-like growth factor-I in patients with proliferative diabetic streptozocin-induced diabetic peripheral neuropathy rats by increasing sciatic
retinopathy, Curr. Eye Res. 23 (2001) 368–371, https://doi.org/10.1076/ nerve IGF-1 and IGF-1R Expression, Front. Pharmacol. 10 (2019) 255, https://doi.
ceyr.23.5.368.5441. org/10.3389/fphar.2019.00255.
[14] F.E. Merimee, T.J. Zapf, J. Insulin-like, growth factors in the fed and fasted states, [39] M. Taleb, Y. Ding, B. Wang, N. Yang, X. Han, C. Du, et al., Dopamine delivery via
J. Clin. Endocrinol. Metab. 55 (2010) 999–1002, https://doi.org/10.1210/jcem- pH-sensitive nanoparticles for tumor blood vessel normalization and an improved
55-5-999. effect of cancer chemotherapeutic drugs, Adv. Health Mater. 9 (18) (2019),
[15] J.L. Wilkinson-berka, C. Wraight, G. Werther, The role of growth hormone, insulin- 1900283, https://doi.org/10.1002/adhm.201900283.
like growth factor and soma- tostatin in diabetic retinopathy, Curr. Med. Chem. 13 [40] C. Sarkar, D. Chakraborty, P.S. Dasgupta, S. Basu, Dopamine is a safe
(2006) 3307–3317, https://doi.org/10.2174/092986706778773086. antiangiogenic drug which can also prevent 5-fluorouracil induced neutropenia,
[16] P. Raman, Ã. Dnb, A.K. Singal, Ã. Ms, A. Behl, Effect of insulin-like growth factor-1 Int J. Cancer 137 (3) (2014) 744–749.
on diabetic retinopathy in pubertal age patients with type 1 diabetes, asia pacific, [41] M.T. Gentile, O. Pastorino, M. Bifulco, L.C. D Amato, HUVEC Tube-formation Assay
J. Ophthalmol. 8 (2019) 319–323, https://doi.org/10.1097/ to Evaluate the Impact of Natural Products on Angiogenesis, J. Vis. Exp. 148
APO.0000000000000250. (2019), https://doi.org/10.3791/58591.
[17] J.F. Payne, V. Tangpricha, J. Cleveland, M.J. Lynn, R. Ray, S.K. Srivastava, Serum [42] G. Wang, J. Liang, L. Gao, Z. Si, X. Zhang, et al., Baicalin administration attenuates
insulin-like growth factor-I in diabetic retinopathy, Mol. Vis. 17 (2011) hyperglycemia-induced malformation of cardiovascular system, Cell Death Dis. 9
2318–2324. (2018) 234, https://doi.org/10.1038/s41419-018-0318-2.
[18] V. Poulaki, A.M. Joussen, N. Mitsiades, C.S. Mitsiades, E.F. Iliaki, A.P. Adamis, [43] M. Lorenzi, C. Gerhardinger, Early cellular and molecular changes induced by
Insulin-Like Growth Factor-I Plays a Pathogenetic Role in Diabetic Retinopathy, diabetes in the retina, Diabetologia 44 (2001) 791–804, https://doi.org/10.1007/
Am. J. Pathol. 165 (2004) 457–469, https://doi.org/10.1016/S0002-9440(10) s001250100544.
63311-1. [44] A.K.W. Lai, A.C.Y. Lo, Animal models of diabetic retinopathy: summary and
[19] A. Pristerà, C. Blomeley, E. Lopes, S. Threlfell, E. Merlini, D. Burdakov, S. Cragg, comparison, J. Diabetes Res. (2013), 106594, https://doi.org/10.1155/2013/
F. Guillemot, S.L. Ang, Dopamine neuron-derived IGF-1 controls dopamine neuron 106594.
firing, skill learning, and exploration, Proc. Natl. Acad. Sci. USA 116 (9) (2019) [45] G. Scuderi, S. Fragiotta, L. Scuderi, C.M. Iodice, A. Perdicchi, Ganglion cell
3817–3826, https://doi.org/10.1073/pnas.1806820116. complex analysis in glaucoma patients: what can it tell us? Eye Brain 12 (2020)
[20] A.E. Ayadi, M.J. Zigmond, A.D. Smith, IGF-1 protects dopamine neurons against 33–44, https://doi.org/10.2147/EB.S226319.
oxidative stress: association with changes in phosphokinases, Exp. Brain Res 234 [46] J. Chhablani, A. Sharma, A. Goud, H.K. Peguda, H.L. Rao, V.U. Begum,
(7) (2016) 1863–1873, https://doi.org/10.1007/s00221-016-4572-1. G. Barteselli, Neurodegeneration in type 2 diabetes: evidence from spectral-domain
[21] A.M. Penha, F. Schaeffel, M. Feldkaemper, Insulin, insulin-like growth factor-1, optical coherence tomography, Investig. Ophthalmol. Vis. Sci. (2015) 6333–6338,
insulin receptor, and insulin-like growth factor-1 receptor expression in the chick https://doi.org/10.1167/iovs.15-17334.
eye and their regulation with imposed myopic or hyperopic defocus, Mol. Vis. 17 [47] Y.J. Choi, J.W. Jeoung, K.H. Park, D.M. Kim, Glaucoma detection ability of
(2011) 1436–1448. ganglion cell-inner plexiform layer thickness by spectral-domain optical coherence
[22] S.R. Panjala, S.A. Thomas, J.J. Steinle, Effects of insulin-like growth factor-1 (IGF- tomography in high myopia, Investig. Ophthalmol. Vis. Sci. (2013) 2296–2304,
1) receptor signaling on rates of apoptosis in retina of dopamine beta hydroxylase https://doi.org/10.1167/iovs.12-10530.
(Dbh-/-) knockout mice, Auton. Neurosci: Basic Clin. 152 (1–2) (2010) 21–26, [48] J.W. Jeoung, Y.J. Choi, K.H. Park, D.M. Kim, Macular ganglion cell imaging study:
https://doi.org/10.1016/j.autneu.2009.08.014. glaucoma diagnostic accuracy of spectral-domain optical coherence tomography,
[23] W.M. Zawada, D.L. Kirschman, J.J. Cohen, K.A. Heidenreich, C.R. Freed, Growth Investig. Ophthalmol. Vis. Sci. (2013), https://doi.org/10.1167/iovs.12-11273.
factors rescue embryonic dopamine neurons from programmed cell death, Exp. [49] S. Bonnin, R. Tadayoni, A. Erginay, P. Massin, Correl. Ganglion Cell Layer.
Neurol. 140 (1) (1996) 60–67, https://doi.org/10.1006/exnr.1996.0115. Thinning Poor Vis. Funct. Resolut. Diabet. Macular Edema (2015), https://doi.org/
[24] S.F. Abcouwer, Angiogenic factors and cytokines in diabetic retinopathy, J. Clin. 10.1167/iovs.14-15503.
Cell. Immunol. 1 (2013) 1–12, https://doi.org/10.4172/2155-9899.Angiogenic. [50] C. Hernández, M. Dal Monte, R. Simó, G. Casini, Neuroprotection as a therapeutic
[25] Y. Dong, C. Qian, G. Wan, A. Schizandrin, Protects human retinal pigment target for diabetic retinopathy, J. Diabetes Res. 2016 (2016), 9508541, https://doi.
epithelial cell line ARPE-19 against HG-Induced cell injury by regulation of miR- org/10.1155/2016/9508541.
145, Mol. Ther. 19 (2020) 42–49, https://doi.org/10.1016/j.omtn.2019.10.026. [51] V. Pérez-Fernández, D.G. Harman, J.W. Morley, M.A. Cameron, Optimized method
[26] W. Wang, A.C.Y. Lo, Diabetic retinopathy: pathophysiology and treatments, Int. J. to quantify dopamine turnover in the mammalian retina, Anal. Chem. 89 (2017)
Mol. Sci. 19 (2018) 1816, https://doi.org/10.3390/ijms19061816. 12276–12283, https://doi.org/10.1021/acs.analchem.7b03216.
[27] C.-Y. Gong, B. Lu, Q.-W. Hu, L.-L. Ji, Streptozotocin induced diabetic retinopathy in [52] C.T. Motz, K.C. Chesler, R.S. Allen, K.L. Bales, L.M. Mees, A.J. Feola, A.Y. Maa, D.
rat and the expression of vascular endothelial growth factor and its receptor, Int. J. E. Olson, P.M. Thule, P.M. Iuvone, A.M. Hendrick, M.T. Pardue, Novel detection
Ophthalmol. 1 (2013) 573–577, https://doi.org/10.3980/j.issn.2222- and restorative levodopa treatment for preclinical diabetic retinopathy, Diabetes
3959.2013.05.03. 69 (2020) 1518–1527, https://doi.org/10.2337/db19-0869.
[28] C.J. Gibson, C.J. Watkins, R.J. Wurtman, Tyrosine administration enhances [53] K. Okimura, Y. Nakane, T. Nishiwaki-Ohkawa, T. Yoshimura, Photoperiodic
dopamine synthesis and release in light-activated rat retina, J. Neural Transm. 160 regulation of dopamine signaling regulates seasonal changes in retinal
(1983) 153–160, https://doi.org/10.1007/BF01243274. photosensitivity in mice, Sci. Rep. 11 (2021) 1843, https://doi.org/10.1038/
[29] P.S. Rao, N. Rujikarn, J.D.H. Tyras, A specific sensitive HPLC method for s41598-021-81540-w.
determination of plasma dopamine, Ch 28 (1989) 307–310, https://doi.org/ [54] G. Ogata, T.W. Stradleigh, G.J. Partida, A.T. Ishida, Dopamine and full-field
10.1007/BF02260781. illumination activate D1 and D2-D5-type receptors in adult rat retinal ganglion
[30] A. Kovács-Valasek, E. Pöstyéni, V. Dénes, A. Mester, G. Sétáló Jr., R. Gábriel, Age- cells, J. Comp. Neurol. 520 (2012) 4032–4049, https://doi.org/10.1002/
related alterations of proteins in albino wistar rat retina, Cells Tissues Organs 210 cne.23159.
(2021) 135–150, https://doi.org/10.1159/000515447. [55] M.L. Firsov, L.A. Astakhova, The role of dopamine in controlling retinal
[31] J. He, H. Wang, Y. Liu, W. Li, D. Kim, H. Huang, Blockade of vascular endothelial photoreceptor function in vertebrates, Neurosci. Behav. Physiol. 46 (2016)
growth factor receptor 1 prevents inflammation and vascular leakage in diabetic 138–145, https://doi.org/10.1007/s11055-015-0210-9.
retinopathy, J. Ophthalmol. 2015 (2015), 605946, https://doi.org/10.1155/2015/ [56] T. Kondo, D. Vicent, K. Suzuma, M. Yanagisawa, G.L. King, M. Holzenberger, C.
605946. R. Kahn, Knockout of insulin and IGF-1 receptors on vascular endothelial cells
[32] C.-Y. Gong, B. Lu, Y.-C. Sheng, Z.-Y. Yu, J.-Y. Zhou, L.-L. Ji, The development of protects against retinal neovascularization, J. Clin. Invest. 111 (2003) 1835–1842,
diabetic retinopathy in goto-kakizaki rat and the expression of angiogenesis-related https://doi.org/10.1172/JCI17455.
signals, Chin. J. Physiol. 59 (2016) 100–108, https://doi.org/10.4077/cjp.2016. [57] J. Ruberte, E. Ayuso, M. Navarro, A. Carretero, V. Nacher, V. Haurigot, M. George,
bae383. C. Llombart, A. Casellas, C. Costa, A. Bosch, F. Bosch, Increased ocular levels of
[33] R. Simó, J.M. Sundstrom, D.A. Antonetti, Ocular Anti-VEGF therapy for diabetic IGF-1 in transgenic mice lead to diabetes-like eye disease, J. Clin. Invest. 113
retinopathy: the role of VEGF in the pathogenesis of diabetic retinopathy, Diabetes (2004) 1149–1157, https://doi.org/10.1172/JCI19478.
Care. 37 (2014), https://doi.org/10.2337/dc13-2002. [58] C. Gerhardinger, K.D. McClure, G. Romeo, F. Podestà, M. Lorenzi, IGF-I mRNA and
[34] T.J. Wubben, Dopamine and early retinal dysfunction in diabetes: insights from a signaling in the diabetic retina, Diabetes 50 (2001), https://doi.org/10.2337/
phase 1 study, Diabetes 69 (2020) 1339–1340, https://doi.org/10.2337/dbi20- diabetes.50.1.175.
0009. [59] D.V. Nguyen, S. Li Calzi, L.C. Shaw, J.L. Kielczewski, H.E. Korah, M.B. Grant, An
[35] H. Zhang, Y. Gao, J. Zhang, K. Wang, T. Jin, H. Wang, K. Ruan, F. Wu, Z. Xu, The ocular view of the IGF-IGFBP system, Growth Horm. IGF Res 23 (2013) 45–52,
effect of total lignans from Fructus Arctii on Streptozotocin-induced diabetic https://doi.org/10.1016/j.ghir.2013.03.001.
retinopathy in Wistar rats, J. Ethnopharmacol. 255 (2020), 112773, https://doi. [60] A.I. Arroba, L. Rodríguez-de la Rosa, S. Murillo-Cuesta, L. Vaquero-Villanueva, J.
org/10.1016/j.jep.2020.112773. M. Hurlé, I. Varela-Nieto, Á.M. Valverde, Autophagy resolves early retinal
[36] K. Chesler, C. Motz, H. Vo, A. Douglass, R.S. Allen, A.J. Feola, M.T. Pardue, inflammation in Igf1-deficient mice, Dis. Model. Mech. 9 (2016) 965–974, https://
Initiation of L-DOPA treatment after detection of diabetes-induced retinal doi.org/10.1242/dmm.026344.
dysfunction reverses retinopathy and provides neuroprotection in rats, Transl. Vis. [61] G. Xi, C. Wai, D. Clemmons, Inhibition of aberrant IGF-I signaling in diabetic male
Sci. Technol. 10 (2021) 8, https://doi.org/10.1167/tvst.10.4.8. rat retina prevents and reverses changes of diabetic retinopathy, J. Diabetes Res.
[37] D. Chakroborty, C. Sarkar, H. Yu, J. Wang, Z. Liu, P. Sarathi, Dopamine stabilizes 2019 (2019), 6456032, https://doi.org/10.1155/2019/6456032.
tumor blood vessels by up-regulating angiopoietin 1 expression in pericytes and

13
S. Upreti et al. Biomedicine & Pharmacotherapy 149 (2022) 112868

[62] Z. Yu, B. Lu, C. Gong, L. Ji, Streptozotocin induced diabetic retinopathy in C 57 [70] P. Wang, X.F. Tian, J.B. Rong, D. Liu, G.G. Yi, Q. Tan, Protein Kinase B (Akt)
mice and the expression of some pro - angiogenic molecules, Int. Eye Sci. 16 (2016) promotes pathological angiogenesis in murine model of oxygen-induced
1–6, https://doi.org/10.3980/j.issn.1672-5123.2016.1.01. retinopathy, Acta Histochem Cytochem 44 (2) (2011) 103–111.
[63] T. Namakachi, A. Matkovits, T. Seki, S. Shioda, Distribution and protective [71] H.-Y.L. Park, J.H. Kim, C.K. Park, Neuronal cell death in the inner retina and the
function of pituitary adenylate cyclase-activating polypeptide in the retina IN influence of vascular endothelial growth factor inhibition in a diabetic rat model,
RETINAL TISSUE, Front. Endocrinol. (Lausanne). 3 (2012) 1–10, https://doi.org/ Am. J. Pathol. 184 (2014) 1752–1762, https://doi.org/10.1016/j.
10.3389/fendo.2012.00145. ajpath.2014.02.016.
[64] Q. Yang, Y. Zhang, J. Jiang, M. Wu, Q. Han, Q. Bo, G. Yu, Y. Ru, X. Liu, M. Huang, [72] X. Zhang, M. Lassila, M.E. Cooper, Z. Cao, Retinal expression of vascular
L. Wang, X. Zhang, J. Fang, X. Li, Protective effects of a novel drug RC28-E endothelial growth factor is mediated by angiotensin type 1 and type 2 receptors,
blocking both VEGF and FGF2 on early diabetic rat retina, Int. J. Ophthalmol. 11 Hypertension 1 (2004) 276–281, https://doi.org/10.1161/01.
(2018) 935–944, https://doi.org/10.18240/ijo.2018.06.07. HYP.0000113628.94574.0f.
[65] N. Gupta, S. Mansoor, A. Sharma, A. Sapkal, J. Sheth, P. Falatoonzadeh, [73] A.N. Witmer, H.G. Blaauwgeers, H.A. Weich, K. Alitalo, G.F.J.M. Vrensen, R.
B. Kuppermann, M. Kenney, Diabetic retinopathy and VEGF, Open Ophthalmol. J. O. Schlingemann, Altered expression patterns of VEGF receptors in human diabetic
7 (2013) 4–10, https://doi.org/10.2174/1874364101307010004. retina and in experimental VEGF-induced retinopathy in monkey, Invest.
[66] L. Moons, C. Vanhole, P. Carmeliet, S. Vanhaesebrouck, H. Danie, N. Intensive, Ophthalmol. Vis. Sci. 43 (2002) 849–857.
C. Unit, Oxygen-Induced Retinopathy in Mice: Amplification by Neonatal IGF-I [74] A. Campos, J. Martins, E.J. Campos, R. Silva, A.F. Ambrósio, Choroidal and retinal
Deficit and Attenuation by IGF-I Administration, Pediatr. Res. 65 (2009) 307–310, structural, cellular and vascular changes in a rat model of Type 2 diabetes, Biomed.
https://doi.org/10.1203/PDR.0b013e3181973dc8. Pharmacother. 132 (2020), 110811, https://doi.org/10.1016/j.
[67] J. Peter, R. Folberg, A. Itin, H. Gnessin, I. Hemo, E. Keshet, Upregulated expression biopha.2020.110811.
of vascular endothelial growth factor in proliferative diabetic retinopathy, 241 LP – [75] A.J. Barber, D.A. Antonetti, T.S. Kern, C.E.N. Reiter, R.S. Soans, J.K. Krady, S.
245, Br. J. Ophthalmol. 80 (1996), https://doi.org/10.1136/bjo.80.3.241. W. Levison, T.W. Gardner, S.K. Bronson, The Ins2 akita mouse as a model of early
[68] P. Kermer, N. Klocker, M. Labes, M. Bahr, Insulin-like growth factor-I protects retinal complications in diabetes, Investig. Ophthalmol. Vis. Sci. (2005)
axotomized rat retinal ganglion cells from secondary death via PI3-K-dependent 2210–2218, https://doi.org/10.1167/iovs.04-1340.
Akt phosphorylation and inhibition of caspase-3 In vivo, J. Neurosci. 20 (2000) [76] L.E.H. Smith, W. Shen, C. Perruzzi, S. Soker, F. Kinose, X. Xu, G. Robinson,
722–728. S. Driver, J. Bischoff, B. Zhang, J.M. Schaeffer, D.R. Senger, Regulation of vascular
[69] F. Qui, h Tong, Y. Wang, J. Tao, H. Wang, L. Chen, Inhibition of miR-21-5p endothelial growth factor-dependent retinal neovascularization by insulin-like
suppresses high glucose-induced proliferation and angiogenesis of human retinal growth factor-1 receptor, Nat. Med. 5 (1999) 1390–1395, https://doi.org/
microvascular endothelial cells by the regulation of AKT and ERK pathways via 10.1038/70963.
maspin, Biosci. Biotechnol. Biochem 82 (8) (2018) 1366–1376, https://doi.org/
10.1080/09168451.2018.1459179.

14

You might also like