• Users Online: 95
  • Print this page
  • Email this page

 
Table of Contents
ORIGINAL ARTICLE
Year : 2023  |  Volume : 66  |  Issue : 1  |  Page : 14-20

GATA-binding protein 4 promotes neuroinflammation and cognitive impairment in Aβ1–42 fibril-infused rats through small nucleolar RNA host gene 1/miR-361-3p axis


1 Department of Internal Medicine-Neurology, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
2 Department of Rheumatology and Immunology, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, China

Date of Submission12-Jul-2022
Date of Decision10-Oct-2022
Date of Acceptance24-Oct-2022
Date of Web Publication20-Feb-2023

Correspondence Address:
Dr. Yanhui Peng
Department of Internal Medicine-Neurology, The Sixth Affiliated Hospital of Xinjiang Medical University, No. 39, Wuxing South Road, Tianshan, Urumqi, Xinjiang Uygur Autonomous Region
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/cjop.CJOP-D-22-00057

Rights and Permissions
  Abstract 


Aging with dysregulated metabolic and immune homeostasis stimulates pyroptosis, neuroinflammation, and cellular senescence, thus contributing to etiopathogenesis of Alzheimer's disease. GATA-binding protein 4 (GATA4) functions as a transcriptional factor in response to DNA damage, and is associated with neuroinflammation and cellular senescence. The role of GATA4 in Alzheimer's disease was investigated. GATA4 was elevated in hippocampus of Aβ1–42 fibril-infused rats. Injection with shRNA targeting GATA4 reduced escape latency with increase of time in target quadrant and number of platform crossings in Aβ1–42 fibril-infused rats. Moreover, knockdown of GATA4 ameliorated morphological changes of hippocampus and reduced amyloid plaque deposition in Aβ1–42 fibril-infused rats. Silence of GATA4 repressed neuroinflammation and apoptosis in Aβ1–42 fibril-infused rats. Loss of GATA4 in Aβ1–42 fibril-infused rats reduced the expression of specificity protein 1 (Sp1) to downregulate long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) and upregulated miR-361-3p. Loss of SNHG1 ameliorated learning and memory impairments in Aβ1–42 fibril-infused rats. Overexpression of Sp1 attenuated GATA4 silence-induced decrease of escape latency, increase of time in target quadrant, and number of platform crossings in Aβ1–42 fibril-infused rats. In conclusion, silence of GATA4 ameliorated cognitive dysfunction and inhibited hippocampal inflammation and cell apoptosis through regulation of Sp1/SNHG1/miR-361-3p.

Keywords: Alzheimer's disease, apoptosis, cognitive impairment, GATA-binding protein 4, miR-361-3p, neuroinflammation, SNHG1, specificity protein 1


How to cite this article:
Liu L, Peng Y, Liu W, Xu J, Li D, Li X. GATA-binding protein 4 promotes neuroinflammation and cognitive impairment in Aβ1–42 fibril-infused rats through small nucleolar RNA host gene 1/miR-361-3p axis. Chin J Physiol 2023;66:14-20

How to cite this URL:
Liu L, Peng Y, Liu W, Xu J, Li D, Li X. GATA-binding protein 4 promotes neuroinflammation and cognitive impairment in Aβ1–42 fibril-infused rats through small nucleolar RNA host gene 1/miR-361-3p axis. Chin J Physiol [serial online] 2023 [cited 2023 Jun 5];66:14-20. Available from: https://www.cjphysiology.org/text.asp?2023/66/1/14/370011




  Introduction Top


Neurodegenerative diseases are characterized by progressive loss of neurons and degeneration, and result in long-term cognitive impairments.[1] Alzheimer's disease refers to a devastating neurodegenerative disease and a leading cause of dementia in aging population.[2] Alzheimer's disease is defined as misfolded accumulation of amyloid beta-peptide (Aβ) and neurofibrillary tangles composed of hyperphosphorylated tau proteins in the brain.[3] Behavioral problems, cognitive deficits, and memory impairment are the clinical manifestations of Alzheimer's disease.[3] Due to the unknown etiology, Alzheimer's disease is devoid of effective treatment.[4] Neuroinflammation exacerbates the accumulation of Aβ and hyperphosphorylated tau proteins, which has been considered to be critical etiopathogenesis of Alzheimer's disease.[4] Targeting neuroinflammation is regarded as functional therapeutic approach to ameliorate the development of Alzheimer's disease.[4]

GATA-binding protein 4 (GATA4) belongs to member of GATA transcription factor family, which is involved in cardiac growth and differentiation.[5] GATA4 was shown to be activated by DNA damage, and triggered senescence through inhibition of autophagy.[6] Decline in autophagy was associated with aging brains, and aging was a risk factor for neurodegenerative disease.[7] Therefore, GATA4 might be involved in pathogenesis of Alzheimer's disease. Moreover, GATA4 has been shown to be upregulated in brains of intracerebral hemorrhage-induced rats, and loss of GATA4 reduced neuronal apoptosis and neurobehavioral scores to attenuate brain damage.[8] GATA4 also promoted inflammation to stimulate cellular senescence.[6] GATA4 might contribute to the development of Alzheimer's disease through promotion of neuroinflammation and neuronal apoptosis.

Long non-coding RNAs (lncRNAs) with regulatory ability at the posttranscriptional and transcriptional levels of target genes,[9] show a prevalent role in pathophysiology of Alzheimer's disease through targeting of miRNAs.[10] LncRNA small nucleolar RNA host gene 1 (SNHG1) was upregulated in Parkinson's disease patients, and promoted neuroinflammation and neuron loss in mouse models with Parkinson's disease.[11] Knockdown of SNHG1 reduced Aβ-induced neuronal injury.[12] GATA4 bind to the promoter region of SNHG1, and SNHG1/GATA4 were involved in the regulation of hypoxia/reoxygenation-induced cardiomyocyte injury.[13] Therefore, GATA4 might be involved in Alzheimer's disease through regulation of SNHG1.

In this study, effects of GATA4 on neuroinflammation, apoptosis, and cognitive impairment in Aβ1–42 fibril-infused rats were investigated, and the underlying mechanism might provide a potential target for Alzheimer's disease.


  Materials and Methods Top


Preparation of Aβ fibrils and viruses

1–42 (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in hexafluoroisopropanol (1 mg/mL; Sigma-Aldrich), and the hexafluoroisopropanol was evaporated using nitrogen gas. Aβ was then vortexed with distilled water: 10X fibril-forming buffer (9:1). The solution was stored at 37°C for 1 week to induce Aβ fibrils according to a previous study.[14] To produce viral vectors, specificity protein 1 (Sp1) sequence, shRNA targeting GATA4 (sh-GATA4), sh-SNHG1, and negative control (sh-NC) (Invitrogen, Carlsbad, CA, USA) were subcloned into pAAV-U6-GFP vector (Cell Biolabs, San Diego, CA, USA). HEK-293 cells were transfected with pAAV-U6-GFP-Sp1 or pAAV-U6-GFP-shRNAs, pAAV-DJ Rep-Cap, and pHelper for 3 days. Cells were then harvested, and the virus was collected using AAV purification kits (Takara Bio, Tokyo, Japan).

Animals

A total of 96 male Sprague-Dawley rats (290–340 g weight and 8 weeks old) were acquired from Vital River Laboratory Animal Technology Company (Beijing, China). Rats were kept in standard conditions, and all the experiments were approved by the Ethics Committee of Xinjiang Medical University (Approval No. SCXK2022-021) and performed in accordance with the National Institutes of Health Laboratory Animal Care and Use Guidelines. Rats were divided into four groups: sham (n = 12), Aβ (n = 12), Aβ+sh-NC (n = 12), Aβ+sh-GATA4 (n = 12), Aβ+sh-SNHG1 (n = 12), Aβ+sh-NC+vector (n = 12), Aβ+sh-GATA4+vector (n = 12), Aβ+sh-GATA4+Sp1 (n = 12). Rats in the sham group were anesthetized with 10% chloral hydrate; 3 μL of sterile saline was delivered bilaterally into the lateral ventricles of rats at coordinates (4.6 mm beneath the dura, 1.5 mm lateral to the sagittal, 1.0 mm anteroposterior from Bregma). Rats in the Aβ groups were also injected with 3 μL of Aβ fibrils once for 3 days according to a previous study.[14] Rats in the Aβ+sh-NC, Aβ+sh-GATA4, Aβ+sh-SNHG1, Aβ+sh-NC+vector, Aβ+sh-GATA4+vector, and Aβ+sh-GATA4+Sp1 were bilaterally infused with 1010 infectious viral particles/mL (1 μL) once to the same spot of Aβ injection 2 weeks before injection with Aβ.

Quantitative reverse transcription-polymerase chain reaction

Three weeks after shRNA infusion, rats were anesthetized and the hippocampus was isolated from each rat (six rats in each group). The tissues were lysed in Trizol (Invitrogen) or miRcute miRNA isolation kit (Tiangen, Beijing, China) to isolate RNAs or total miRNAs, respectively. The RNAs were then reverse-transcribed into cDNAs. The cDNAs were subjected to quantitative reverse transcription polymerase chain reaction analysis with SYBR Green Master (Roche, Mannheim, Germany). GAPDH (forward: 5'-GGATTTGGTCGTATTGGG-3' and reverse: 5'-GGAAGATGGTGATGGGATT-3') and U6 (forward: 5'-CTCGCTTCGGCAGCACA-3' and reverse: 5'-AACGCTTCACGAATTTGCGT-3') served as endogenous controls, and the expression of SNHG1 (forward: 5'-CCGCTCGAGATTTAGGTGACACTATAGAAGTTCTCATTTTTCTACTGCTCG-3' and reverse: 5'-ATAGTTTAGCGGCCGCTTTTTTTTTTTTTTTTATGTAATCAATCATTTTAT-3'), tumor necrosis factor-alpha (TNF-α) (forward: 5'-ACTGAACTTCGGGGTGATTG-3' and reverse: 5'-GCTTGGTGGTTTGCTACGAC-3'), interleukin (IL)-1β (forward: 5'-CACCTTCTTTTCCTTCATCTTTG-3' and reverse: 5'-GTCGTTGCTTGTCTCTCCTTGTA-3'), IL-6 (forward: 5'-TGATGGATGCTTCCAAACTG-3' and reverse: 5'-GAGCATTGGAAGTTGGGGTA-3'), and miR-361-3p (forward: 5'-ACACTCCAGCTGGGTCCCCCAGGTGTGATTC-3' and reverse: 5'-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGAAATCAGA-3') were determined by 2−ΔΔCT method.

Western blot

Hippocampal tissues were lysed in radioimmunoprecipitation assay buffer with protease inhibitor cocktail (Beyotime, Beijing, China). Protein samples were segregated using sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and then transferred onto nitrocellulose membranes. Membranes were blocked in 5% dry milk, and then incubated with primary antibodies: anti-GATA4 and anti-GAPDH (1:2000), anti-BAX and anti-BCL-2 (1:3000), and anti-Sp1 and 6E10 (1:4000). The membranes were then incubated with secondary antibodies (1:5000), and subjected to chemiluminescence reagent kit (Beyotime). All the antibodies were purchased from Abcam (Cambridge, MA, USA).

Enzyme-linked immunosorbent assay

Lysates from hippocampal tissues were harvested postcentrifugation at 12,000 ×g for 1 h. Levels of TNF-α, IL-1β, and IL-6 were detected by enzyme-linked immunosorbent assay kits (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China).

Pathological analysis

Hippocampal tissues were fixed in 10% formalin and then embedded in paraffin. The tissues were sliced serially into 5 μm sections. The sections were dewaxed with xylene and hydrated in gradient concentrations of alcohol. Sections were then stained with hematoxylin and eosin (Sigma-Aldrich). The morphological changes of hippocampus were observed under microscope (Olympus, Tokyo, Japan). The sections were also treated with Gill's hematoxylin solution (Sigma-Aldrich), and then incubated with alkaline sodium chloride solution. Sections were stained with Congo Red working solution (Sigma-Aldrich) before microscopic examination.

Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining

Hippocampal sections were implemented at 90°C for antigen retrieval, and treated with Protease K (Sigma-Aldrich). Sections were suspended in TDT buffer (Sigma-Aldrich), and incubated overnight with anti-serum alkaline phosphatase and anti-digoxin complex (Sigma-Aldrich). Following counterstaining with DAPI, sections were observed under microscope to determine apoptosis in hippocampus. The apoptotic ratio was quantified using Image-Pro Plus.

Morris water maze test

Three weeks after shRNA infusion, rats in each group (six rats in each group) were trained in a water-filled tub (height 90 cm and diameter 160 cm). An escape platform (height 25 cm and diameter 12 cm) was located in 1 cm below the water surface. At the beginning of each trial, rats were placed in the water maze from one of four different coordinates (N, S, E, and W), and then guided to reach the platform in 1 min. Rats were subjected to four trials per day for 5 consecutive days. The time to find the platform with 1 min was recorded as escape latency. On day 6, the platform was removed for spatial probe test. The swim paths of rats were recorded via ANY-maze video tracking system (Stoelting, Wood Dale, IL, USA); number of platform crossings and time in quadrant were recorded in 2 min.

Statistical analysis

All the data were expressed as mean ± standard error of mean and analyzed by Student's t-test or one-way analysis of variance. P < 0.05 was considered statistically significant.


  Results Top


Silence of GATA-binding protein 4 alleviated learning and memory impairments in Aβ1–42 fibril-infused rats

To establish Alzheimer's disease, rats were infused with Aβ fibrils. GATA4 was elevated in hippocampus of Aβ-infused rats [Figure 1]a. Rats were injected with sh-GATA4 virus, and then subjected to Aβ infusion. Injection with sh-GATA4 virus reduced the expression of GATA4 [Figure 1]a. Escape latency of rats was increased by Aβ infusion [Figure 1]b. However, knockdown of GATA4 decreased the escape latency of Aβ-infused rats [Figure 1]b. Aβ infusion induced cognitive deficits as characterized by the swim paths [Figure 1]c, while knockdown of GATA4 ameliorated the cognitive deficits in Aβ-infused rats [Figure 1]c. Knockdown of GATA4 also attenuated Aβ-induced decrease of number of platform crossings [Figure 1]d and time in target quadrant [Figure 1]e in rats, suggesting the protective effect of GATA4 silence against learning and memory impairments in Aβ1–42 fibril-infused rats.
Figure 1: Silence of GATA4 alleviated learning and memory impairments in Aβ1–42 fibril-infused rats. (a) GATA4 was elevated in hippocampus of Aβ-infused rats, and injection with sh-GATA4 virus reduced the expression of GATA4. (b) Injection with sh-GATA4 virus decreased escape latency of Aβ-infused rats. (c) Representative images of the swim paths of rats on day 6 in different groups. Black dot was the location of the platform. (d) Injection with sh-GATA4 virus increased number of platform crossings of Aβ-infused rats on day 6 of spatial probe test. (e) Injection with sh-GATA4 virus increased time in target quadrant of Aβ-infused rats on day 6 of spatial probe test. *** versus sham, P < 0.001. ##, ### versus Aβ + sh-NC, P < 0.01, P < 0.001. GATA4: GATA-binding protein 4, sh-GATA4: shRNA targeting GATA4, sh-NC: negative control.

Click here to view


Silence of GATA-binding protein 4 alleviated morphological changes in Aβ1–42 fibril-infused rats

The hippocampal tissues were then subjected to pathological analysis via hematoxylin and eosin and Congo red staining. Results showed that neurons in Aβ-infused rats showed disordered, rupture in nuclei, shrink in volume, and enlarged intercellular space [Figure 2]a. Moreover, the number of neurons was decreased [Figure 2]a. However, injection with sh-GATA4 virus ameliorated the pathological changes in hippocampus through increase of neurons [Figure 2]a. Congo red staining showed that knockdown of GATA4 reduced amyloid plaque deposition in hippocampus of Aβ-infused rats [Figure 2]b. Western blot analysis also showed the upregulation of anti-amyloid beta (6E10) in hippocampal tissues of Aβ-infused rats [Figure 2]c. Knockdown of GATA4 reduced the expression of 6E10 [Figure 2]c.
Figure 2: Silence of GATA4 alleviated morphological changes in Aβ1–42 fibril-infused rats. (a) Injection with sh-GATA4 virus ameliorated the pathological changes in hippocampus of Aβ-infused rats through increase of neurons. N = 6. (b) Injection with sh-GATA4 virus reduced amyloid plaques deposition in hippocampus of Aβ-infused rats. (c) Knockdown of GATA4 reduced anti-amyloid beta (6E10) in hippocampal tissues of Aβ-infused rats. N = 6. *** versus sham, P < 0.001. ### versus Aβ + sh-NC, P < 0.001. GATA4: GATA-binding protein 4, sh-GATA4: shRNA targeting GATA4, sh-NC: negative control.

Click here to view


Silence of GATA-binding protein 4 alleviated secretion of inflammatory factors in Aβ1–42 fibril-infused rats

Levels of TNF-α, IL-1β, and IL-6 were increased in hippocampus of Aβ-infused rats [Figure 3]a. Silence of GATA4 exerted an anti-inflammatory effect against Aβ-infused rats through downregulation of TNF-α, IL-1β, and IL-6 [Figure 3]b.
Figure 3: Silence of GATA4 alleviated secretion of inflammatory factors in Aβ1–42 fibril-infused rats. (a) Injection with sh-GATA4 virus reduced TNF-α, IL-1β, and IL-6 mRNAs in hippocampus of Aβ-infused rats. (b) Injection with sh-GATA4 virus reduced TNF-α, IL-1β, and IL-6 in hippocampus of Aβ-infused rats. *** versus sham, P < 0.001. ##, ### versus Aβ + sh-NC, P < 0.01, P < 0.001. GATA4: GATA-binding protein 4, sh-GATA4: shRNA targeting GATA4, TNF-α: Tumor necrosis factor-alpha, IL: Interleukin, sh-NC: negative control.

Click here to view


Silence of GATA-binding protein 4 alleviated hippocampal apoptosis in Aβ1–42 fibril-infused rats

The number of TUNEL-positive cells in hippocampus of rats was increased by Aβ infusion [Figure 4]a. However, knockdown of GATA4 reduced the number of TUNEL-positive cells to inhibit apoptosis in hippocampal tissues of Aβ-infused rats [Figure 4]a. Moreover, loss of GATA4 attenuated the Aβ-induced decrease of Bcl-2 and increase of Bax in the hippocampus of rats [Figure 4]b.
Figure 4: Silence of GATA4 alleviated hippocampal apoptosis in Aβ1–42 fibril-infused rats. (a) Injection with sh-GATA4 virus reduced the number of TUNEL-positive cells in Aβ-infused rats. (b) Injection with sh-GATA4 virus increased protein expression of Bcl-2 and decreased Bax in the hippocampus of Aβ-infused rats. *** versus sham, P < 0.001. ### versus Aβ + sh-NC, P < 0.001. GATA4: GATA4-binding protein 4, sh-GATA4: shRNA targeting GATA4, sh-NC: negative control.

Click here to view


GATA-binding protein 4 regulated small nucleolar RNA host gene 1/miR-361-3p in Aβ1–42 fibril-infused rats

Protein expression of Sp1 was enhanced in hippocampus of Aβ-infused rats, while reduced by silence of GATA4 [Figure 5]a. Loss of GATA4 attenuated Aβ-induced decrease of miR-361-3p and increase of SNHG1 in the hippocampus of rats [Figure 5]b. Moreover, injection with Sp1 virus decreased the expression of miR-361-3p, increased Sp1 and SNHG1 in hippocampus of Aβ-infused rats [Figure 5]c. Loss of SNHG1 reduced the expression of SNHG1 and enhanced miR-361-3p [Figure 5]c. Knockdown of SNHG1 decreased escape latency and increased number of platform crossings and time in target quadrant in Aβ-infused rats [Figure 5]d. Overexpression of Sp1 attenuated GATA4 deficiency-induced decrease of escape latency, increase of number of platform crossings, and time in target quadrant in Aβ-infused rats [Figure 5]e, demonstrating that GATA4 might regulate Sp1/SNHG1/miR-361-3p axis to protect against Alzheimer's disease.
Figure 5: GATA4 regulated SNHG1/miR-361-3p in Aβ1–42 fibril-infused rats. (a) Injection with sh-GATA4 virus reduced protein expression of Sp1 in Aβ-infused rats. (b) Injection with sh-GATA4 virus reduced SNHG1 expression and enhanced miR-361-3p in Aβ-infused rats. (c) Injection with Sp1 virus decreased the expression of miR-361-3p and increased Sp1 and SNHG1 in hippocampus of Aβ-infused rats. Loss of SNHG1 reduced the expression of SNHG1 and enhanced miR-361-3p. (d) Knockdown of SNHG1 decreased escape latency and increased number of platform crossings and time in target quadrant in Aβ-infused rats. (e) Overexpression of Sp1 attenuated GATA4 deficiency-induced decrease of escape latency, increase of number of platform crossings, and time in target quadrant in Aβ-infused rats. *** versus sham, P < 0.001. ##, ### versus Aβ + sh-NC or Aβ + sh-GATA4, P < 0.01, P < 0.001. GATA4: GATA-binding protein 4, SNHG1: Small nucleolar RNA host gene 1, sh-GATA4: shRNA targeting GATA4, Sp1: Specificity protein 1, sh-NC: negative control.

Click here to view



  Discussion Top


GATA transcription factors recruit chromatin remodeling proteins and co-regulators to regulate gene expression involved in development of urogenital, cardiovascular, and hematopoietic systems, livers, and brains.[15] GATA4 has been shown to be involved in inflammation and senescence of pyramidal neurons,[6] as well as neuronal apoptosis of rats with intracerebral hemorrhage.[8] This study found that GATA4 was also associated with cognitive impairments, neuroinflammation, and neuronal apoptosis of Aβ1–42 fibril-infused rats.

Senile plaques, neurofibrillary tangles, and lipid granule accumulation are the three defining neuropathological features of cerebral cortex in patients with Alzheimer's disease.[16] Presence of amyloid plaques within the brain is a pathological hallmark of Alzheimer's disease, and intracerebroventricular injection of Aβ fibrils stimulated endogenous Aβ aggregation, tangle formation, and hyperphosphorylation of tau, thus resulting in neurotoxicity and dementia.[14] Therefore, Aβ-infused rats were widely used as in vivo model of Alzheimer's disease.[14] In this study, Aβ fibrils also triggered learning and memory impairments in rats with increase of escape latency, decrease of number of platform crossings, and time in target quadrant. GATA4 was elevated in hippocampus of Aβ-infused rats, and silence of GATA4 ameliorated learning and memory impairments and alleviated morphological changes and amyloid plaque deposition in hippocampus of Aβ-infused rats.

Age-related stimuli, such as gut microbiota dysbiosis, metainflammation, cellular senescence, oxi-inflammation, mitochondrial dysfunction, and defective autophagy, promote the secretion of pro-inflammatory cytokines to induce neuroinflammation and neuronal apoptosis, and contribute to the development of Alzheimer's disease.[4] TNF-α, IL-1β, and IL-6 were produced by activated microglia to induce neuron damage.[17] Anti-neuroinflammatory agents were used in clinical trials for Alzheimer's disease.[18] GATA4 functioned as an upstream regulator of NF-κB signaling to mediate the expression of inflammatory factors.[6] Moreover, GATA4 also promoted neuronal apoptosis in rats with intracerebral hemorrhage.[8] This study showed that silence of GATA4 reduced the release of TNF-α, IL-1β, and IL-6 in Aβ-infused rats, and suppressed the apoptosis of hippocampal tissues. Oxidative stress stimulated loss of synapses and neurons, tau hyperphosphorylation, and Aβ deposition, and antioxidants were regarded as potential strategies for Alzheimer's disease.[19] Excessive reactive oxygen species promoted the activation of GATA4, and GATA4 modulated the effects of oxidative stress on differentiation of cardiomyogenesis.[20] Therefore, GATA4 might also promote oxidative stress in Aβ1–42 fibril-infused rats.

GATA4 bind to the promoter of Sp1 and induced the gene expression of Sp1.[21] Sp1 was dysregulated in frontal cortex of brains in Alzheimer's disease.[22] Sp1 was regulated by inflammatory factor, IL-1β, and mediated the expression of Alzheimer's disease-related proteins, such as tau and amyloid precursor protein.[22] Sp1 promoted neuronal apoptosis through activation of AMPK signaling in patients with Alzheimer's disease.[23] Silence of GATA4 in this study reduced the expression of Sp1 to attenuate Alzheimer's disease. Sp1 induced the activation of SNHG1.[24] SNHG1 was elevated in brains of patients with Parkinson's disease, and promoted neuroinflammation in lipopolysaccharide-induced BV2 cells.[25] Moreover, knockdown of SNHG1 increased the expression of miR-361-3p to reduce Aβ-induced neuronal injury in Alzheimer's disease.[12] Results in this study also demonstrated that silence of GATA4 reduced the expression of SNGH1 and enhanced miR-361-3p in hippocampus of Aβ-infused rats. Therefore, GATA4 might contribute to the development of Alzheimer's disease through regulation of Sp1/SNHG1/miR-361-3p axis.


  Conclusion Top


In sum, loss of GATA4 exerted anti-inflammatory and anti-apoptotic effects on Aβ-infused rats, and alleviated learning and memory impairments in Aβ1–42 fbril-infused rats. Silence of GATA4 reduced Sp1 and SNHG1 to enhance miR-361-3p in Aβ1–42 fbril-infused rats. However, the role of GATA4/Sp1/SNHG1/miR-361-3p in in vitro and in vivo models of Alzheimer's disease should be investigated in further research.

Financial support and sponsorship

This work was supported by the Natural Science Foundation of Xinjiang Uygur Autonomous Region (Grant No. 2021D01C456).

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Fang P, Kazmi SA, Jameson KG, Hsiao EY. The microbiome as a modifier of neurodegenerative disease risk. Cell Host Microbe 2020;28:201-22.  Back to cited text no. 1
    
2.
Peng L, Bestard-Lorigados I, Song W. The synapse as a treatment avenue for Alzheimer's disease. Mol Psychiatry 2022;27:2940-9.  Back to cited text no. 2
    
3.
Yin X, Qiu Y, Zhao C, Zhou Z, Bao J, Qian W. The role of amyloid-beta and tau in the early pathogenesis of Alzheimer's disease. Med Sci Monit 2021;27:e933084.  Back to cited text no. 3
    
4.
Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol 2015;14:388-405.  Back to cited text no. 4
    
5.
Garg V, Kathiriya IS, Barnes R, Schluterman MK, King IN, Butler CA, et al. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature 2003;424:443-7.  Back to cited text no. 5
    
6.
Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, et al. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science 2015;349:aaa5612.  Back to cited text no. 6
    
7.
Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol 2019;15:565-81.  Back to cited text no. 7
    
8.
Xu H, Cao J, Xu J, Li H, Shen H, Li X, et al. GATA-4 regulates neuronal apoptosis after intracerebral hemorrhage via the NF-κB/Bax/caspase-3 pathway both in vivo and in vitro. Exp Neurol 2019;315:21-31.  Back to cited text no. 8
    
9.
Yu XP, Liu CG, Qiu F, Xing F, Han SJ, Han Y, et al. Breast cancer overall-survival can be predicted with a 19 lncRNA tissue signature. Eur J Gynaecol Oncol 2021;42:838-43.  Back to cited text no. 9
    
10.
Sabaie H, Amirinejad N, Asadi MR, Jalaiei A, Daneshmandpour Y, Rezaei O, et al. Molecular insight into the therapeutic potential of long non-coding RNA-associated competing endogenous RNA axes in Alzheimer's disease: A systematic scoping review. Front Aging Neurosci 2021;13:742242.  Back to cited text no. 10
    
11.
Zhao X, Gu C, Wang Y. PAD4 selective inhibitor TDFA protects lipopolysaccharide-induced acute lung injury by modulating nuclear p65 localization in epithelial cells. Int Immunopharmacol 2020;88:106923.  Back to cited text no. 11
    
12.
Gao Y, Zhang N, Lv C, Li N, Li X, Li W. lncRNA snhg1 knockdown alleviates amyloid-β-induced neuronal injury by regulating ZNF217 via sponging miR-361-3p in Alzheimer's disease. J Alzheimers Dis 2020;77:85-98.  Back to cited text no. 12
    
13.
Gong J, Dou L, Zhou Y. Positive feedback loop of lncRNA SNHG1/miR-16-5p/GATA4 in the regulation of hypoxia/reoxygenation-induced cardiomyocyte injury. Mol Med Rep 2022;25:28.  Back to cited text no. 13
    
14.
Sheng C, Xu P, Liu X, Peng W, Xiang D, Luo S. Bushen-Tiansui formula improves cognitive functions in an Aβ 1-42 fibril-infused rat model of Alzheimer's disease. Neural Plast 2020;2020:8874885.  Back to cited text no. 14
    
15.
Tremblay M, Sanchez-Ferras O, Bouchard M. GATA transcription factors in development and disease. Development 2018;145:dev164384.  Back to cited text no. 15
    
16.
Price JL, editor. Tangles and plaques in healthy aging and Alzheimer's disease: Independence or interaction? Seminars in Neuroscience. 1994;6:395-442.  Back to cited text no. 16
    
17.
Babić Leko M, Nikolac Perković M, Klepac N, Štrac DŠ, Boroveči F, Pivac N, et al. IL-1β, IL-6, IL-10, and TNFα single nucleotide polymorphisms in human influence the susceptibility to Alzheimer's disease pathology. J Alzheimers Dis 2020;75:1029-47.  Back to cited text no. 17
    
18.
Zhou W, Hu W. Anti-neuroinflammatory agents for the treatment of Alzheimer's disease. Future Med Chem 2013;5:1559-71.  Back to cited text no. 18
    
19.
Chen Z, Zhong C. Oxidative stress in Alzheimer's disease. Neurosci Bull 2014;30:271-81.  Back to cited text no. 19
    
20.
Li T, Zhang X, Jiang K, Liu J, Liu Z. Dural effects of oxidative stress on cardiomyogenesis via Gata4 transcription and protein ubiquitination. Cell Death Dis 2018;9:246.  Back to cited text no. 20
    
21.
Zhang C, Zhao H, Song X, Wang J, Zhao S, Deng H, et al. Transcription factor GATA4 drives RNA polymerase III-directed transcription and transformed cell proliferation through a filamin A/GATA4/SP1 pathway. J Biol Chem 2022;298:101581.  Back to cited text no. 21
    
22.
Citron BA, Dennis JS, Zeitlin RS, Echeverria V. Transcription factor Sp1 dysregulation in Alzheimer's disease. J Neurosci Res 2008;86:2499-504.  Back to cited text no. 22
    
23.
Cao Y, Tan X, Lu Q, Huang K, Tang X, He Z. miR-590-3 and SP1 promote neuronal apoptosis in patients with Alzheimer's disease via AMPK signaling pathway. Contrast Media Mol Imaging 2021;2021:6010362.  Back to cited text no. 23
    
24.
Zhao MW, Qiu WJ, Yang P. SP1 activated-lncRNA SNHG1 mediates the development of epilepsy via miR-154-5p/TLR5 axis. Epilepsy Res 2020;168:106476.  Back to cited text no. 24
    
25.
Cao B, Wang T, Qu Q, Kang T, Yang Q. Long noncoding RNA SNHG1 promotes neuroinflammation in Parkinson's disease via regulating miR-7/NLRP3 pathway. Neuroscience 2018;388:118-27.  Back to cited text no. 25
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Materials and Me...
Results
Discussion
Conclusion
References
Article Figures

 Article Access Statistics
    Viewed1215    
    Printed50    
    Emailed0    
    PDF Downloaded210    
    Comments [Add]    

Recommend this journal