Skip to main content
Neural Regeneration Research logoLink to Neural Regeneration Research
. 2023 Jan 5;18(9):2037–2046. doi: 10.4103/1673-5374.366493

Single-nuclei RNA sequencing uncovers heterogenous transcriptional signatures in Parkinson’s disease associated with nuclear receptor-related factor 1 defect

Piniel Alphayo Kambey 1,2, Wen-Ya Liu 1, Jiao Wu 1, Bakwatanisa Bosco 3, Iqra Nadeem 1, Kouminin Kanwore 1, Dian-Shuai Gao 1,*
PMCID: PMC10233763  PMID: 36926730

Abstract

graphic file with name NRR-18-2037-g001.jpg

Previous studies have found that deficiency in nuclear receptor-related factor 1 (Nurr1), which participates in the development, differentiation, survival, and degeneration of dopaminergic neurons, is associated with Parkinson’s disease, but the mechanism of action is perplexing. Here, we first ascertained the repercussion of knocking down Nurr1 by performing liquid chromatography coupled with tandem mass spectrometry. We found that 231 genes were highly expressed in dopaminergic neurons with Nurr1 deficiency, 14 of which were linked to the Parkinson’s disease pathway based on Kyoto Encyclopedia of Genes and Genomes analysis. To better understand how Nurr1 deficiency autonomously invokes the decline of dopaminergic neurons and elicits Parkinson’s disease symptoms, we performed single-nuclei RNA sequencing in a Nurr1 LV-shRNA mouse model. The results revealed cellular heterogeneity in the substantia nigra and a number of activated genes, the preponderance of which encode components of the major histocompatibility II complex. Cd74, H2-Ab1, H2-Aa, H2-Eb1, Lyz2, Mrc1, Slc6a3, Slc47a1, Ms4a4b, and Ptprc2 were the top 10 differentially expressed genes. Immunofluorescence staining showed that, after Nurr1 knockdown, the number of CD74-immunoreactive cells in mouse brain tissue was markedly increased. In addition, Cd74 expression was increased in a mouse model of Parkinson’s disease induced by treatment with 6-hydroxydopamine. Taken together, our results suggest that Nurr1 deficiency results in an increase in Cd74 expression, thereby leading to the destruction of dopaminergic neurons. These findings provide a potential therapeutic target for the treatment of Parkinson’s disease.

Key Words: 6-hydroxydopamine, dopaminergic neurons, dopamine transporter, nuclear receptor-related factor 1, Parkinson’s disease, proteomics analysis, Seurat clustering, single-nuclei RNA sequencing, substantia nigra, tyrosine hydroxylase

Introduction

Ten out of 107 individuals with hereditary Parkinson’s disease (PD) were reported to have two mutations in exon 1 of the nuclear receptor-related factor 1 (Nurr1) gene (Le et al., 2003). Nurr1 (also referred to as Nr4a2) is a member of the nuclear receptor superfamily and belongs to the Nr4a subgroup. Nur77 (Nr4a1) and Nor1 (Nr4a3) are the other two members of the Nr4a subgroup, and their functions appear to differ markedly from that of Nurr1 (Maxwell and Muscat, 2006; Chen et al., 2020b). Multiple studies have shown that Nurr1 is required for the formation, division, preservation, and longevity of midbrain dopaminergic neurons, which are most impaired in the brains of individuals with PD (Kadkhodaei et al., 2009; Decressac et al., 2013; Rodríguez-Traver et al., 2016).

Nurr1 appears to function as a transcriptional factor in the synthesis of classical markers of dopaminergic neurons, including tyrosine hydroxylase (TH), dopamine transporter (DAT), aromatic amino acid decarboxylase (AADC), and vesicular monoamine transporter (VMAT) (Jankovic et al., 2005; Kadkhodaei et al., 2009; Dong et al., 2016). The age-related decrease in Nurr1 expression in midbrain dopaminergic neurons has attracted researchers’ attention because it could explain the increased morbidity of PD in the elderly (Chu et al., 2002).

Furthermore, Nurr1 immunoreactivity is considerably lower in the substantia nigra of individuals who suffer from PD compared with healthy individuals (Balu et al., 2008). Multiple studies have shown that Nurr1 is expressed at lower levels in postmortem brain samples (Chu et al., 2006) and peripheral blood samples (Liu et al., 2012; Lou and Liao, 2012; Montarolo et al., 2016; Ruiz-Sánchez et al., 2017; Li et al., 2018; Yang et al., 2019) from patients with PD. In line with these clinical findings, genetic ablation of Nurr1 in mice results in a complete loss of dopaminergic neurons in the substantia nigra and ventral tegmental region, potentially leading to the death of newborn mice (Zetterström et al., 1997). Several dopaminergic neuron markers that are critical for the neurons of the nigrostriatal trajectory, such as TH, DAT, and AADC, are absent in Nurr1-deficient animals (Chu et al., 2006; Kadkhodaei et al., 2009). Moreover, heterozygous Nurr1-deficient animals exhibit greater vulnerability to the toxins 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (Le et al., 1999). Nurr1 conditional knockout mice mimic the early symptoms of PD, displaying a series of neuropathological changes such as a decrease in the number of striatal dopaminergic neurons and motor impairment, suggesting that they could be a useful model for studying PD (Kadkhodaei et al., 2013).

With recent technological advancements, single-cell/-nuclei RNA sequencing has emerged as a revolutionary technology that, in addition to cellular characterization (Papalexi and Satija, 2018; Abdelgawad et al., 2021) within specific microenvironments (Tikhonova et al., 2019; Gong et al., 2021), is critical in determining disease mechanisms (Chen et al., 2020c; Kim et al., 2021) as well as in drug discovery (Wu et al., 2017; Chen et al., 2020a) and drug repurposing (Xu et al., 2021). Here, we employed single-nuclei RNA sequencing to investigate how Nurr1 deficiency alone leads to dopaminergic neuron degeneration in PD.

Methods

Cell culture and Nurr1_shRNA lentiviral transient transfection

The immortalized dopaminergic MES23.5 cell line (RRID: CVCL_J351, Chinese Academy of Sciences, Shanghai, China), authenticated by short tandem repeat loci, was maintained at a density of 5 × 104 cells/mL in 25 cm2 dishes (Corning, Acton, MA, USA) pre-coated with 25 mg/mL poly-D-lysine. In an incubator with 5% CO2 at 37°C, cells were grown in Dulbecco’s modified Eagle medium/Nutrient Mixture F-12 (DMEM/F12) containing 10% fetal bovine serum (Atlas, Fort Collins, CO, USA) and 1% penicillin (Invitrogen, Carlsbad, CA, USA). Cells were grown to a minimum of 80% confluency before being passaged and were passaged 10 to 15 times to reach full maturity before being used in subsequent experiments. The mature cells were plated at a density of 5 × 104 cells/cm2 on poly-L-lysine-coated six-well plates 24 hours before lentiviral transient transfection and then switched to serum-free DMEM/F12 (Gibco, Shanghai, China) overnight. Lipofectamine 2000 (Invitrogen) was used to carry out the transfections following the manufacturer’s instructions. Three different plasmids (LV-Nr4a2-mus-526, LV-Nr4a2-mus-667, and LV-Nr4a2-mus-787) (Genepharma, Suzhou, China) were used, and the one with the greatest knockdown effect was used for further experiments. Additional file 1 (735.7KB, pdf) explains how the transfection was conducted; for the sake of simplicity, only the most effective knockdown plasmid (Genepharma) is shown. Briefly, after cells were grown in a six-well plate, they were fasted overnight by switching to serum-free DMEM/F12 (2 mL in each well). Subsequently, the lentivirus (50 μL) was added to every 1 mL of serum-free medium, according to the respective groups. This was followed by an immediate addition of lipofectamine (5 μL). The proportion of cells that exhibited green fluorescence as detected by a fluorescence microscope (BX51, Olympus, Tokyo, Japan) 72 hours after transfection was 90%. To screen for successfully transfected cells, the transfection mixture was removed and replaced with a fresh complete medium containing 8 μg/mL puromycin (ThermoFisher Scientific, Shanghai, China). Transfected cells were subsequently grown in puromycin-containing medium to maintain the plasmid.

Quantitative polymerase chain reaction

Briefly, each dish of cells was washed with phosphate-buffered saline (PBS, Dalian Meilun Biotechnology, Dalian, China), and 1 mL of TRIzol was added (Ambion-Life Science Technologies, Shanghai, China). Then, quantitative polymerase chain reaction was performed as we described previously (Kambey et al., 2021b; Additional file 1 (735.7KB, pdf) ). The primers used were NR4A2 forward: 5′-GTG TTC AGG CGC AGT ATG G-3′, reverse: 5′-TGT ATT CTC CCG AAG AGT GGT AA-3′, and β-actin forward: 5′-ACG GCC AGG TCA TCA CTA TTG-3′, reverse: 5′-CAA GAA GGA AGG CTG GAA AAG A-3′.

Mass spectrometry

Immunoprecipitation

To determine the effects of Nurr1 knockdown on neuron proteomics, we cultured MES23.5 cells and then divided them into two groups (Nurr1 LV_shRNA and Nurr1 LV_Scramble), with three replicates each. Samples were gathered, and protein extraction was carried out as described earlier (Kaboord and Perr, 2008). Because the level of Nurr1 enrichment in dopaminergic neurons appeared to be modest, we opted to undertake immunoprecipitation using magnetic beads (Medchemexpress, Shanghai, China) prior to performing tandem mass spectrometry (MS). Total protein was isolated from the samples and immunoprecipitated with a Nurr1 antibody (rabbit, 1:200, Abcam, Shanghai, China, Cat# ab41917, RRID: AB_776887) before trypsin digestion and processing for label-free quantification tandem MS, which are described in detail below.

Trypsin digestion

After washing the samples repeatedly with 200 μL of 1× PBS and centrifuging them at 12,000 × g for 10 minutes at 4°C, the supernatant was discarded. The samples were then resuspended in 100 μL of 50 mM NH4HCO3 (Tianjin Chengyuan Chemical, Tianjin, China). Disulfide bonds were reduced by adding dithiothreitol (Chongqing Borning Chemical & Industrial, Chongqing, China) to a final concentration of 10 mM, and the mixture was incubated at 56°C for 1 hour. Iodoacetamide solution (Sigma, Shanghai, China) was added to final concentration of 50 mM, and the reaction was allowed to proceed for 40 minutes. Trypsin (Sigma, Shanghai, China) was added to the protein substrate at a mass ratio of 1:100, and the reaction was allowed to proceed at 37°C for 4 hours. Then, trypsin was added once more at a mass ratio of 1:100, and the mixture was digested for 16 hours at 37°C. The peptide was desalted using a self-filling desalting column after enzymatic digestion and dried in a vacuum centrifuge concentrator at 45°C. After dissolving the peptides in sample solution containing 0.1% formic acid and 2% acetonitrile (Sigma, Shanghai, China), the mixture was centrifuged at 4°C for 10 minutes with a full oscillating vortex at 13,200 × g. The supernatants were then transferred to fresh sample tubes for MS analysis.

Label-free liquid chromatography coupled with tandem MS (LC-MS/MS)

High-pH reverse performance liquid chromatography was used to separate the peptides on an Agilent 300 Extend C18 column (Agilent Technologies, Santa Clara, CA, USA) with a particle size of 5 μm, an internal diameter of 4.6 mm, and a length of 250 mm. The process involved the following steps: At a pH value of 9, the peptide gradient ranged from 8% to 32% acetonitrile. Sixty components were separated over 60 minutes; the peptides were then concatenated into 14 components, which were dehydrated using a vacuum dryer to prepare for the following steps. After being dispersed in 0.1% formic acid (solvent A), the tryptic peptides were deposited directly onto a pre-column phase and then separated using a reversed-phase analytical column (Acclaim PepMap RSLC, Thermofisher Scientific, Waltham, MA, USA). The gradient consisted of a 26-minute increase from 6% to 23% solvent B (0.1% formic acid in 98% acetonitrile, Thermofisher Scientific, Waltham, MA, USA), an 8-minute increase from 23% to 35%, a 3-minute climb to 80%, and a final 3-minute hold at 80% (on an EASY-nLC 1000 ultra-performance liquid chromatography system at a constant flow rate of 500 nL/min). The peptides were subjected to nanospray ionization followed by ultra-performance liquid chromatography tandem mass spectrometry (MS/MS) using an internet-connected Q ExactiveTM Plus (Thermofisher Scientific, Waltham, MA, USA). The electrospray voltage used was 2.0 kV. For a comprehensive scan, the m/z range used was 350 to 1800, and whole peptides were identified at a resolution of 70,000 using an Orbitrap mass spectrometer (Thermofisher Scientific, Waltham, MA, USA). After the peptides were selected for MS/MS with a normalized collision energy value of 28, the fragments were identified by the Orbitrap spectrometer at a resolution of 17,500. A data-dependent approach in which one MS scan was followed by 20 MS/MS scans with 15.0-second dynamic exclusion was used. Automatic gain control was set at 5E4, and 100 m/z was assigned to the fixed initial mass.

LC-MS/MS data analysis

The MaxQuant search engine v1.5.2.8 (Max Planck Institute, Berlin, Germany) was used to process and analyze the data. The UniProt database (UniProt Consortium, 2021; Wang et al., 2021) was combined with a reverse decoy database to analyze the tandem MS/MS spectra. The other parameters were set and the final analysis was performed as described previously (Yu et al., 2021). The Clue gene ontology (ClueGO) plug-in for Cytoscape (Cytoscape Consortium, San Diego, CA, USA) was used to identify differentially expressed genes that may be associated with PD.

Animals

Owing to hormonal influences and the low body weight of female mice (Fillingim and Ness, 2000; Craft et al., 2004; Mogil and Chanda, 2005), only male mice were used in this study. Sixty-five C57BL/6J male mice aged 8 weeks, weighing 25–30 g, were supplied by the Animal Center of Xuzhou Medical University (license No. SYXK [Su] 2020-0048). For the first phase of the animal experiments, mice were divided into two groups: Nurr1 knockdown (Nurr1 LV_shRNA, n = 30) and control (Nurr1 LV_Scramble, n = 20). In the second phase of the animal experiments (n = 15), Nurr1 LV_shRNA was injected into the left side of the striatum (0.5 mm anterior- posterior, 1.5 mm lateral, 3.5 mm ventral to the dural surface), while Nurr1 LV_Scramble was injected into the right side as the control (same brain coordinates). Animals (five mice in each cage) were provided with consistent meals and free access to water and maintained under a 12/12-hour light/dark cycle with controlled temperature (15–28°C), and humidity (40–65%) conditions. Animals were treated in accordance with the rules set by the Ethical Committee for the Use of Research Animals of Xuzhou Medical University (approval No. L20210226270, June 16, 2021). All experiments were designed and reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines (Percie du Sert et al., 2020).

Stereotaxic injection of Nurr1 LV_shRNA

Nurr1 LV_shRNA viral stock comprising 1 × 106 infectious viral units in a 200 μL volume was supplied by Santa Cruz Biotechnology (Dallas, TX, USA). These lentiviral particles, intended for use in gene silencing, were delivered as transduction-ready viral particles. The particles contained three to five expression constructs comprising a target-specific 19–25 nucleotide sequences plus hairpin yielding an shRNA designed to silence gene expression. Additional Figure 1 (77.9KB, pdf) shows how the Nurr1_shRNA plasmid was produced. After general anesthesia was induced by intraperitoneal injection of 45 mg/kg sodium pentobarbital (Sigma, St. Louis, MO, USA), mice were positioned within a stereotaxic apparatus (RWD, San Diego, CA, USA), and the bite bar was adjusted to a distance of 0 mm. After exposing the skull, a high-speed dental drill was used to create the burr hole. Twenty animals were injected with 7 μL of Nurr1 LV_shRNA in the left striatum (0.5 mm anterior- posterior, 1.5 mm lateral, 3.5 mm ventral to the dural surface (Earls et al., 2019). The control group (20 mice) received 7 μL of Nurr1 LV_Scramble (Santa Cruz Biotechnology, Dallas, TX, USA). The injection was delivered over 4 minutes using a Hamilton micro syringe with a capacity of 10 μL. The needle was then left in place for an additional 4 minutes, as described in a prior study (Aoi et al., 2000). TH immunoreactivity was used to confirm the degeneration of dopamine neurons after 4 weeks. To validate the activated genes after single nuclei-RNA sequencing, a second phase of lentiviral infusion (Nurr1 LV_shRNA and Nurr1 LV_Scramble, n = 10) was performed, with the contralateral striatum used as a control.

Validation of activated genes identified by single-nuclei RNA sequencing in the PD mouse model

We established a PD mouse model as we described previously (Kambey et al., 2021b). In brief, 6-OHDA (Medchemexpress) was prepared (5 μg in 1 μL of 0.9% normal saline containing 0.2% ascorbic acid). Then, 2 μL of 6-OHDA was injected into the left striatum, and 2 μL of normal saline was injected into the right striatum of the same mouse to serve as a control (n = 15). The same brain coordinates were used for each injection, as described in more detail above. Four weeks later, quantitative polymerase chain reaction (qPCR) was performed. Briefly, the midbrain was extracted on ice and rapidly homogenized in 1 mL of TRIzol. The subsequent steps are shown in Additional file 1 (735.7KB, pdf) . The delta-delta Ct technique was used to analyze the results, and glyceraldehyde-3-phosphate dehydrogenase (Gapdh) mRNA was used as an internal reference. The following primers were used as: Gapdh, forward: 5′-CGG GGT CCC AGC TTA GGT TC-3′, reverse: 5′-GCC CAA TAC GGC CAA ATC CG-3′; Cd74, forward: 5′-AGT GCG ACG AGA ACG GTA AC-3′, reverse: 5′-CGT TGG GGA ACA CAC ACC A-3′.

Behavioral testing

Open field test

In the first week after stereotaxic surgery, six mice died (five from the Nurr1 knockdown group and one from the control group). The remaining animals (n = 34) were separated into two groups (control versus experimental) prior to performing the behavioral tests. To assess locomotion (in the mice that only received lentiviral infusion, in phase one of the animal experiments), the open field test was performed 4 weeks post-stereotaxic surgery. Locomotion analysis is one of the most well-established tests in rodents and is also the most standardized method for measuring motor function (Schwarting et al., 1993; Zhang et al., 2020). It can be carried out in an open or closed field. Mice were placed in the center of a 50 cm × 50 cm enclosure with 50 cm-high walls and given 10 minutes to explore. The center section was lit at 300 lx, and the amount of time spent in the central zone over 30 minutes was recorded. Having all four paws in the middle area was classified as entry into the central zone. The ANY-maze video-tracking system (Muromachi Kikai, Tokyo, Japan) was used to digitally record and analyze the animals’ behavior.

Elevated body swing test

Three weeks after lentiviral infusion/stereotaxic surgery (only for phase one of the animal experiments), the elevated body swing test was performed for both groups (Nurr1 knockdown and control). The same test was repeated in the fourth and fifth weeks. The elevated body swing test is also referred to as the tail suspension test (Can et al., 2012), especially in studies conducted in mice to investigate antidepressants, in which immobility time rather than swing path direction is assessed. The same mice that were assessed in the open field test were subjected to the elevated body swing test. The elevated body swing test was carried out according to Borlongan & Sanberg’s original description (Borlongan and Sanberg, 1995) with minimal modifications. Each mouse was placed in a transparent cage measuring 36 cm × 19.5 cm × 18.5 cm and permitted to choose a comfortable position with all four of its paws on the ground. It was then elevated to around 10 cm from the bottom of the cage by the tip of the tail, and the direction of the first body swing was recorded. A body swing was defined as bending the upper body away from the vertical axis by more than 10° to either side. The animal was then replaced on the ground and allowed to adjust its position in preparation for the subsequent swing test. Each mouse was re-suspended once it was clearly balanced and showed no predilection for one side or the other. The test was carried out 20 times for each animal. If the animal did not start swinging, the base of its tail was gently tapped to encourage it to do so. To prevent affecting the direction of swings, the assessor alternated the hand used to pick up the mouse and where he stood in relation to the testing cage. Additionally, the test was carried out in an area that was free of any objects that could influence swing direction. The fraction of swings toward the right side was calculated. The number of initial head or upper body turns contralateral to the lesioned side was determined and standardized over 20 consecutive trials as follows: percentage of contralateral recovery = [1 – (lateral turning in 20 trials – 10)/10] × 100 (Borlongan and Sanberg, 1995; Henderson et al., 1999; Chang et al., 2003).

Western blot assay

Four weeks post-lentiviral infusion, western blotting was performed for dopaminergic neuron markers. First, 300 μL of a lysis buffer cocktail comprising radioimmunoprecipitation assay buffer (Medchemexpress) and phenylmethanesulfonyl fluoride inhibitor (Medchemexpress) in a 100:1 ratio was prepared and added to ice-cold cells or midbrain samples. The samples were homogenized for 30 minutes with vortexing every 5 minutes. The homogenates were then centrifuged at 4°C at 14,000 × g for 30 minutes. The supernatants were transferred to fresh 0.5 mL tubes and maintained on ice, and the pellets were discarded. A bicinchoninic acid kit (Medchemexpress) was used to determine the protein concentrations of the samples following the manufacturer’s instructions. The optical densities of the protein standards and samples were calculated. The protein concentrations of the samples were standardized using radioimmunoprecipitation assay buffer, and the samples were then boiled in loading buffer (a reducing agent) at 100°C for 10 minutes. Next, 20-μg protein samples were subjected to electrophoresis on a 10% polyacrylamide gel at a constant voltage of 80 V while the proteins migrated through the stacking gel, at which point the voltage was increased to 100 or 120 V for separation of the protein bands. The proteins were then electrotransferred (100 V, 1 hour) to nitrocellulose membranes. The membranes for 2 hours in Tris-buffered saline before incubation with primary antibodies to TH (rabbit, 1:500, Proteintech, Shanghai, China, Cat# 25859-1-AP, RRID: AB_2716568), DAT (mouse, 1:500, Novus Biologicals, Shanghai, China, Cat# H00006531-A01, RRID:AB_670572), glial cell line-derived neurotrophic factor (GDNF) (rabbit, 1:1000, Abcam, Cat# ab18956, RRID:AB_2111379), Nurr1/NR4A2 (rabbit, 1:500, Proteintech, Cat# 10975-2-AP, RRID:AB_2153760), and β-actin (mouse, 1:2000, Santa Cruz Biotechnology, Shanghai, China, Cat# sc-47778 HRP, RRID:AB_2714189) overnight at 4°C. The membranes were then incubated for 6 hours at room temperature (25–32°C) and subsequently probed with appropriate secondary antibodies (goat anti-rabbit IgG H&L (HRP), 1:200, Abcam, Cat# ab6721, RRID: AB_955447, and goat anti-mouse IgG H&L (HRP), 1:200, Abcam, Cat# ab6789, RRID: AB_955439). Following the blotting process, the membranes were examined using a scanner (LI-COR Biotechnology, Lincoln, NE, USA) and an image analyzer (LI-COR Biotechnology, Lincoln, Nebraska, USA). The optical density of bands relative to β-actin bands was analyzed with ImageJ software (v1.52a, National Institute of Health, Bethesda, MD, USA) (Schneider et al., 2012).

Immunohistochemistry

Four weeks post-lentiviral infusion, immunohistochemistry was conducted as we described previously (Kambey et al., 2021b). The detailed protocol is provided in Additional file 1 (735.7KB, pdf) .

Immunofluorescence staining

Four weeks post-lentiviral infusion, immunofluorescence staining was performed. Following pentobarbital (45 mg/kg) anesthesia and cardiac perfusion (with 100 mL of PBS followed by 150 mL of cold 4% paraformaldehyde), the brains were harvested and post-fixed in 4% paraformaldehyde for an additional day before being cryoprotected in a solution of 20% (for 24 hours) and 30% (for 24 hours) sucrose in 0.1 M phosphate buffer at 4°C. Coronal brain sections (including the substantia nigra and other midbrain areas) were made at a thickness of 16 μm and air-dried overnight. The sections were then blocked for 2 hours at room temperature with 0.1% Triton X-100 and 10% normal goat serum diluted in 0.1 M phosphate buffer. TH and Nurr1 were detected by incubating sections with TH antibody (rabbit, 1:500, Proteintech, Cat# 25859-1-AP, RRID: AB_2716568 or mouse, 1:400, Santa Cruz Biotechnology, Dallas, TX, USA, Cat# SC-25269, RRID: AB_628422), and Nurr1/NR4A2 polyclonal antibody (rabbit, 1:500, Proteintech, Cat# 10975-2-AP, RRID: AB_2153760) overnight at 4°C. After being thoroughly washed, the stained sections were incubated for 6 hours at room temperature with Alexa Fluor 488-conjugated AffiniPure goat anti-rabbit IgG (1:200, Jackson ImmunoResearch, Carlsbad, CA, USA, Cat# 111-545-003, RRID: AB_2338046). For double labeling for TH and CD74 or Nurr1 and CD74, midbrain sections were incubated overnight at 4°C with antibodies to TH (1:500) and CD74 (mouse, 1:500, Proteintech, Cat# 66390-1-Ig, RRID: AB_2881766) or Nurr1 (1:500) and CD74 (1:500). The slices were then probed for 6 hours at room temperature with Alexa Fluor 488-conjugated AffiniPure goat anti-rabbit IgG (1:200, Jackson ImmunoResearch Carlsbad, CA, USA, Cat# 111-545-003, RRID: AB_2338046) and Alexa Fluor 594-conjugated AffiniPure goat anti-mouse IgG (1:200, Jackson ImmunoResearch Carlsbad, CA, USA, Cat# 128-585-003, RRID: AB_2783778). 4′,6-Diamidino-2-phenylindole (DAPI) nuclear counterstain was applied for 15 minutes. The slides were mounted and examined under a fluorescence microscope. The immunoreactivity relative to the counterstain (DAPI) was assessed and reported.

Single-nuclei RNA sequencing

Nuclear isolation

Four weeks post-lentiviral infusion, the midbrain was harvested from five mice in each group for nuclear isolation. Nuclei were isolated using a slightly revised 10× Genomics® protocol (10× Genomics, Shanghai, China). Briefly, cold lysis buffer containing 10 mM Tris-HCl, 10 mM sodium chloride, 3 mM MgCl2 and 0.1% Nonidet P40 (10× Genomics, Shanghai, China) was used to break up the tissues. Then, the suspension was filtered and centrifuged, and the supernatant was discarded. The pellets containing the nuclei were washed in “nuclei wash and resuspension buffer” containing 1× PBS, 1% bovine serum albumin, and 0.2 U/μL RNase inhibitor (10× Genomics, Shanghai, China), then filtered and centrifuged a second time. To isolate the nuclei, the pellets were first suspended in DAPI solution for 10 minutes. Then, a FACS Diva Cell Sorter (BD Biosciences, San Jose, CA, USA) was used to isolate single nuclei that were positive for DAPI.

Library generation and sequencing

A Chromium Next GEM Single Cell 3’ Kit v3.1 (10× Genomics, Pleasanton, CA, USA) was used to construct complementary DNA libraries from isolated nuclei. An Agilent 2100 Bioanalyzer System (Agilent Technologies) was used to assess complementary DNA quality, and sequencing was performed on an Illumina NovaSeq 6000-S2 apparatus (Illumina, San Diego, CA, USA).

Transcript quantification, filtration, and processing

To produce FASTQ files from the raw base call outputs, the mkfastq pipeline in Cell Ranger (10× Genomics, Pleasanton, USA) was used. Cell Ranger count pipeline v.3.0 (10× Genomics, Pleasanton, USA) was used to build a gene-barcode unique molecular identifier (UMI) count matrix for each sample. We only included genes in the analysis that had at least one UMI count, and cells with fewer than 400 genes or with a mitochondrial UMI count greater than 10% were excluded. Ribosomal and mitochondrial genes were excluded. The processed UMI feature-barcode matrices constructed with Cell Ranger v2.3.4 (10× Genomics, Pleasanton, USA) were analyzed with the Seurat package using R software (Robert Gentleman and Ross Ihaka, Boston, MA, USA) (Satija et al., 2015; Hao et al., 2021).

Normalization, sample integration, and cell clustering

Samples were standardized using the “LogNormalize” technique (Booeshaghi and Pachter, 2021; Seth et al., 2022) to reduce cell-specific biases prior to downstream analyses. To perform multi-sample data integration, we used the weighted-nearest neighbor approach (Hao et al., 2021). This is the Satija task force’s most recent single-cell sequencing data integration technique for combining numerous single-cell samples. Using the Louvain algorithm, the normalized data were aggregated. UMAP (Uniform Manifold Approximation and Projection), a new fluid learning technique for dimensional reduction that consists of theoretical frameworks based on Riemann geometry and algebraic topology, was used to depict the cell clustering results. Using Seurat’s FindAllMarkers tool (https://www.rdocumentation.org/packages/Seurat/versions/4.0.5/topics/FindAllMarkers), we identified marker genes for each cluster.

SingleR (https://www.bioconductor.org/packages/release/bioc/html/SingleR.html), which is a novel computational technique for unbiased single-cell ribonucleic acid sequencing (scRNA-seq) cell type recognition that uses reference transcriptome datasets of pure cell types to determine the cell from which each individual cell originated, was used to annotate cell types (Mabbott et al., 2013; Grün et al., 2016; La Manno et al., 2016; Aran et al., 2019). Even when automatic cell annotation is employed, it is frequently necessary to perform manual annotation due to the inconsistencies that exist in the marker datasets and single-cell atlases that automated techniques rely on (Clarke et al., 2021). Thus, we supplemented the automated annotation with known cell markers based on the Labome (Tanapat, 2013), CellMarker (Zhang et al., 2019), and PanglaoDB (Franzén et al., 2019) databases.

Differential gene expression

A negative binomial test with a false discovery rate adjusted P-value of < 0.05 was performed in Seurat 4.0 (Rahul Satija, Lower Manhattan, NY, USA) to identify genes with a log fold enrichment of 0.25 that were found in at least 25% of the cluster’s cells. Briefly, the following parameters were taken into account: the proportion of genes expressed in the cell population, pct.1 or pct.2 > 0.25; expression multiple, avg log2FC > 0.25; and P val adj < 0.01, where pct.1 or pct.2 denotes the percentage of cells in which the gene was found in the first or second group, respectively. In this study, the Nurr1 LV_shRNA group and control group clusters were determined by differential marker gene expression analysis (FindAllMarkers feature in Seurat), and differentially expressed genes between each cluster were then identified.

Functional analysis

Using Singleseqgset (https://github.com/arc85/singleseqgset) in Seurat, the differentially expressed genes were analyzed for gene ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and Reactome enrichment. The stratified GO layout was visualized with directed acyclic graphs (Aranguren et al., 2007; Textor et al., 2016). Singleseqgset is a package used to perform scRNA-seq data gene set enrichment analysis by applying a fundamental underlying statistics to calculate the enrichment of gene sets of interest across clusters (Zou et al., 2021).

Statistical analysis

Although sample sizes were not predetermined using statistical methods, our sample sizes are comparable to those reported in earlier publications (Zhang et al., 2022; Zhou et al., 2022). The analysis did not omit any animals or data points, and the assessors were not aware of the group assignments. GraphPad Prism version 9.0.0 for Windows (GraphPad Software, San Diego, CA, USA, www.graphpad.com) was employed for statistical analysis. Tukey’s multiple comparisons test preceded by one-way analysis of variance was used to compare multiple groups, whereas Student’s t-test was used for comparisons between two groups. The data are presented as the mean ± standard deviation (SD). P < 0.05 was deemed statistically significant.

Results

Nurr1 knockdown reduces TH expression

TH is a well-known dopaminergic neuron marker (Mendez et al., 2005; Tian et al., 2008). To determine whether Nurr1 is necessary for dopaminergic neuron survival, TH expression was analyzed after Nurr1 knockdown. First, we sought to identify the plasmid that conferred the greatest level of knockdown. MES23.5 cells were transiently transfected with the three plasmids using Lipofectamine™ 2000. Fluorescence was evident after 72 hours, indicating that the transfection was successful. Additional file 1 (735.7KB, pdf) explains how the transfection was conducted. For the sake of simplicity, only the most effective knockdown plasmid is shown. The knockdown efficiency of the plasmids was also determined by qPCR, which showed that LV-Nr4a2-mus-526 conferred the greatest level of knockdown (Figure 1A and B). The western blot results indicated that Nurr1 expression was significantly lower in cells transfected with LV-Nr4a2-mus-526 compared with the control (P = 0.0487, t = 4.366, df = 2; Figure 1C and D). TH expression was markedly lower in Nurr1 knockdown cells compared with control cells (P = 0.0021, t = 21.95, df = 2; Figure 1E and F).

Figure 1.

Figure 1

Nurr1 knockdown reduces TH expression.

(A) Nurr1 (Nr4a2) knockdown in a MES23.5 cells as assessed by fluorescence microscopy (original magnification 4×, scale bars: 200 μm). The green fluorescence indicates Nurr1 expression. (B) The plasmid that induced the greatest degree of knockdown was determined by quantitative polymerase chain reaction. (C, D) Nurr1 expression was significantly reduced in the knockdown cells compared with the control group. (E, F) Tyrosine hydroxylase expression was dramatically reduced after Nurr1 knockdown. The data are presented as the mean ± SD (n = 4). *P < 0.05, **P < 0.01 (one-way analysis of variance followed by Tukey’s multiple comparisons). LV-NC: Lentiviral-normal control; LV-Nr4a2-mus: lentiviral, Nr4a2 promoter, mouse; TH: tyrosine hydroxylase.

LC-MS/MS reveals up-regulated genes linked with PD

To identify changes in protein expression associated with Nurr1 knockdown and to determine whether these changes are associated with PD, label-free quantitative LC-MS/MS proteomics analysis was carried out. A log2-transformed fold change (log2FC) of 1 or –1 and a false discovery rate value of 0.05 were used to define differentially expressed proteins. We found that 231 genes were upregulated, but no genes were downregulated, in Nurr1 knockdown cells compared with the control group (Additional Table 1 (376.4KB, pdf) ). Using ClueGO, a Cytoscape plug-in that generates functionally structured GO category networks by combining GO terms with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, we identified 14 differentially expressed genes (Ndufa9, Ndufa4, Ndufa2, Psmd12, Gnai3, Tubb4b, Sdhb, Psma6, Psmc3, Ndufs3, Uqcrfs1, Vdac1, Ndufv1, and Slc25a4) that were linked to the PD pathway (Figure 2). The majority of these genes are involved in key biological processes (Additional Figure 2 (118.1KB, pdf) ).

Figure 2.

Figure 2

Immunoprecipitation followed by label-free quantitative LC-MS/MS proteomics analysis identified up-regulated genes linked with Parkinson’s disease.

(A) KEGG pathway analysis results showing various genes that are linked to Parkinson’s disease. (B) The names of the genes, as well as their P- and Q-values. The blue box highlights those associated with Parkinson’s disease. (C) ClueGO analysis revealed a Parkinson’s disease node. Nodes of the same color indicate related clusters. ClueGO: Clue Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; LC-MS/MS: liquid chromatography coupled with tandem mass spectrometry.

Pathological effects of Nurr1 knockdown in a mouse model

Next, we first performed in vivo behavioral analyses to determine whether Nurr1 knockdown recapitulates behavioral features of PD. The results indicated that there was a marked difference between the Nurr1 knockdown and control groups in the open field test. Unlike the control group, the Nurr1 knockdown group showed a significant difference between the number of clockwise and anticlockwise rotations (P = 0.0041, t = 3.287, df = 18; Figure 3A and B). In addition, animals in the Nurr1 knockdown group crossed the line fewer times than animals in the control group (P = 0.0006, t = 4.140, df = 18; Figure 3A and C). The elevated body swing test demonstrated a significantly lower degree of contralateral turning recovery in the Nurr1 knockdown group compared with the control group (P = 0.0001, t = 22.31, df = 12; Figure 3D).

Figure 3.

Figure 3

Behavioral analysis revealed marked alteration of locomotion and contralateral turning in Nurr1 knockdown mice relative to the control group.

(A) The two groups were compared in an open field test. (B) Mice in the Nurr1 knockdown group turned primarily to the side with the most dopaminergic neurons. (C) Analysis of line crossings in the open field test revealed a significant difference between the two groups. (D) In the elevated body swing test there was a marked decrease in the proportion of contralateral turning recovery in the Nurr1 knockdown group compared with the control group. The data are presented as the mean ± SD (n = 12). **P < 0.01, ***P < 0.001 (Student’s t-test). Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA.

The methods used to assess TH and Nurr1 immunoreactivity in the striatum and perform double labeling of TH and Nurr1 in the substantia nigra are described in Additional file 1 (735.7KB, pdf) . The immunofluorescence staining results showed a noticeable decrease in Nurr1+ and TH+ neurons in the substantia nigra, as indicated in Figure 4A and B, respectively, suggesting that Nurr1 knockdown leads to dopaminergic neurons degeneration.

Figure 4.

Figure 4

Nurr1 (A) and tyrosine hydroxylase (B) immunoreactivity.

There was a noticeable decrease in the number of Nurr1+ and TH+ neurons in the substantia nigra of the Nurr1 knockdown group relative to the control group. Scale bars: 200 μm (upper), 100 μm (lower). Nurr1: Nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; TH: tyrosine hydroxylase.

The decline in dopaminergic neuron numbers was assessed by western blotting of midbrain samples using dopaminergic neuron markers (TH and DAT). GDNF expression was also ascertained because it is regulated by Nurr1 (Decressac et al., 2012). In comparison to the control group, there was a significant reduction in the expression of these proteins in the Nurr1 knockdown group [TH (P = 0.0275, t = 3.392, df = 4), DAT (P = 0.0059, t = 5.336, df = 4), Nurr1 (P = 0.0344, t = 3.153, df = 4), GDNF (P = 0.0119, t = 4.373, df = 4)] (Figure 5AE).

Figure 5.

Figure 5

Western blot assay reveals a decline in dopaminergic neuron marker expression in the Nurr1 knockdown group compared with the control group.

(A) Immunoblotting for Nurr1, GDNF, and two markers of dopaminergic neurons (TH and DAT). (B–E) Quantitative analysis. The data are presented as a mean ± SD (n = 3). *P < 0.05, **P < 0.01 (Student’s t-test). DAT: Dopamine transporter; GDNF: glial cell line-derived neurotrophic factor; Nurr1: nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; TH: tyrosine hydroxylase.

Cellular heterogeneity in the substantia nigra following Nurr1 knockdown

To better understand how the Nurr1 deficiency alone causes a decline in dopaminergic neuron numbers and elicits PD-like symptoms, we performed single-nuclei RNA sequencing. We first established an animal model of PD by stereotactically injecting shRNA lentiviral particles into the left striatum. Four weeks later, dopamine neuron degeneration and Parkinsonism were confirmed by behavioral testing, western blotting, and immunohistochemistry. Midbrain samples were collected as we described earlier (Kambey et al., 2021b). Samples from five mice from the control group and five mice from the Nurr1 LV-shRNA group were enzymatically digested into single-cell suspensions before being sequenced individually on the 10× Genomics platform. Only genes that had at least one UMI count detected in at least one cell were included in the subsequent analysis. Cells that had fewer than 400 genes or more than 10% mitochondrial unique molecular identifier counts were excluded. After filtering the cells using the lowest and highest thresholds for nUMI, nGene, and mitochondrial RNA genes, 16,093 and 15,260 cells were obtained for the control and Nurr1 LV-shRNA groups, respectively. Similar results were obtained by comparing nUMI, nGene, and mitochondrial mRNA percentages (pMito) (Additional Figure 3 (97.8KB, pdf) ). Batch effects among the samples were noticed and corrected. To normalize expression levels, we used the “LogNormalize” tool of the “Normalization” function, and we decreased variables by principal component analysis to reduce the dimensions of the analysis. Figure 6A shows the total number of cells in each cluster. Figure 6B depicts the distribution of cells on UMAP in each group. We used the Seurat feature of integrating data analyses from multiple conditions to categorize the cell types in all of the samples; that is, by using UMAP’s dimensionality reduction algorithm in conjunction with unsupervised cell clustering (Figure 6C). On the basis of the independently expressed unique transcriptional patterns, 22 cell clusters were identified using the Seurat clustering tool (Figure 6C). Differential gene expression was assessed for each cluster using the Wilcox algorithm to identify marker genes (Figure 6D). The 22 clusters were annotated into six clusters-dopaminergic neurons, GABAergic neurons, glutamatergic neurons, microglia, oligodendrocytes, and astrocytes-using SingleR supplemented with manual annotation. SingleR is a cutting-edge computational technique for impartial cell type detection in scRNA-seq that uses baseline transcriptomic datasets of pure cell types to separately identify the cell of origin of each individual cell (Aran et al., 2019; Figure 6E and F).

Figure 6.

Figure 6

Cellular heterogeneity in the substantia nigra following silencing of the transcription factor Nurr1.

(A) The LogNormalize tool of the “Normalization” function was used to standardize gene expression values. The lower graph shows the total number of cells in each cluster. (B) UMAP plot showing clusters in the Nurr1 LV_shRNA and Nurr1 LV_scramble groups (the numbers 0–22 represent individual clusters). (C) The two groups were integrated for further analysis. (D) The “findallmarkers” tool in Seurat was used to identify the marker genes in each cluster. The numbers indicate the total number of marker genes identified in each cluster. (E) SingleR was used to automatically identify cell types, and manual annotation was used to augment the results. The results correspond to the UMAP diagram. (F) Distribution of the annotated clusters. Nurr1: Nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; UMAP: Uniform Manifold Approximation and Projection.

Single-nuclei RNA sequencing identified genes that are activated in the context of Nurr1 knockdown, the majority of which encode components of major histocompatibility II complex, suggesting a mechanistic basis for the role of Nurr1 deficiency in PD

Next, we sought to identify markers that were highly expressed across all clusters. Genes enriched in each cluster were identified using the Seurat package “FindAllMarkers” (Additional Table 2 (4MB, pdf) ). Our analysis identified a number of genes previously not known to be associated with PD, the majority of which encode components of major histocompatibility complex II and were expressed at high levels in Nurr1 LV-shRNA samples. Cd74, H2-Ab1, H2-Aa, H2-Eb1, Lyz2, Mrc1, Slc47a1, Ms4a4b, and Ptprc2 were found in the oligodendrocyte cluster, and Slc6a3 was found in the dopaminergic neuron cluster. Table 1 shows the top 20 differentially expressed marker genes. A violin plot depicts the expression of these marker genes throughout the 22 cell clusters (Figure 7A and B). Figure 7C depicts the expression of these marker genes in several cell clusters using a UMAP approach. Among the top 10 genes, four interacting nodes were detected utilizing the “STRING” tool (Figure 7D). The majority of these genes are associated with the major histocompatibility complex II and encode proteins involved in antigen processing and presentation. According to SingleR analysis, which was augmented with manual cell annotation in Seurat, all of the top 10 markers identified for all of the clusters were overexpressed in the oligodendrocyte cluster (Table 1). For each of the five most significantly differentially expressed marker genes, we created a heatmap showing the levels of gene expression (Figure 7E). When pathway annotations were examined, the most significantly enriched pathway among all expressed marker genes was antigen processing and presentation (Figure 7F and Additional Table 3). Other pathways included cell adhesion molecules, allograft rejection, graft-versus-host disease, and many others. On the Gene Ontology Directed Acyclic Graph, most of the nodes are related to antigen processing and presentation (Figure 7G). GO biological process annotation of all marker genes found that the majority of the genes are involved in peptide antigen processing and presentation, antigen processing and presentation, and immune system processes, among other aspects (Additional Table 4 (1.9MB, pdf) ).

Table 1.

The top 20 differentially expressed marker genes

Gene p_val avg_log2FC pct.1 pct.2 p_val_adj Cluster
Cd74 0 5.279959 0.552 0.029 0 Oligodendrocytes
H2-Ab1 0 4.792272 0.471 0.013 0 Oligodendrocytes
H2-Aa 0 4.378477 0.471 0.016 0 Oligodendrocytes
Ms4a4b 0 4.358793 0.804 0.005 0 Oligodendrocytes
H2-Eb1 0 4.281058 0.448 0.013 0 Oligodendrocytes
Slc47a1 0 4.211992 0.717 0.004 0 Oligodendrocytes
Ptprc2 0 3.967357 0.75 0.021 0 Oligodendrocytes
Lyz2 0 3.83447 0.557 0.021 0 Oligodendrocytes
Slc6a3 0 3.822566 0.686 0.014 0 Dopamine neurons
Mrc1 0 3.809226 0.557 0.005 0 Oligodendrocytes
H2-K12 1.78E-187 3.80541 0.783 0.064 3.82E-183 Microglia
Flt1 0 3.802803 0.687 0.026 0 Oligodendrocytes
Slc18a2 0 3.694368 0.843 0.051 0 GABAergic neurons
Slc38a23 1.53E-86 3.620089 0.861 0.407 3.27E-82 Oligodendrocytes
Ptgds3 6.33E-139 3.547415 0.969 0.823 1.36E-134 Astrocytes
Slc7a111 0 3.523647 0.597 0.04 0 Astrocytes
H2-Q7 0 3.517585 0.652 0.009 0 Oligodendrocytes
Slco1a4 0 3.40731 0.582 0.018 0 Oligodendrocytes
F630028O10Rik 0 3.342438 0.505 0.002 0 Oligodendrocytes
Dcn 0 3.310526 0.468 0.026 0 Oligodendrocytes

pct.1 means the percentage of cells expressing the gene in group 1 (Nurr1 LV_shRNA). Pct.2 is the percentage of cells expressing the gene in group 2 (Nurr1 LV_Scramble).

Figure 7.

Figure 7

Single-nuclei RNA sequencing revealed activation of genes encoding components of major histocompatibility II complex, suggesting a possible mechanism for Nurr1 deficiency-related defects in Parkinson’s disease.

(A) Violin plot showing expression of the top 10 marker genes in the 22 cell clusters. (B) Top 10 marker genes shown by the cluster in which they were expressed. (C) UMAP plots displaying expression of the top 10 marker genes in each separate cell cluster, with each gene’s expression level represented by a different color. (D) Interaction of the top 10 marker genes as deduced using the STRING tool. (E) Gene expression heatmap for the five most significant marker genes identified from the 22 clusters. (F) KEGG pathway annotation of the 20 most significant marker genes. The horizontal coordinate is Rich factor, the size of the dot represents the number of different genes that are enriched in the term, and the color represents the P-value of the path. (G) A Gene Ontology Directed Acyclic Graph (DAG) constructed in R was used to infer/visualize the hierarchical structure of the gene ontology. Most of the nodes are related to antigen processing and presentation. The figure shows the 10 most significant GO-BP terms. UMAP: Uniform Manifold Approximation and Projection; KEGG: Kyoto Encyclopedia of Genes and Genomes; GO-BP: Gene Ontology-Biological Processes.

Additional Table 3.

Pathway’s annotation

Category PathwayID Pathway List Total Pvalue adjustPvalue Gene
Organismal Systems mmu04612 Antigen processing and presentation 9 90 3.08E-11 3.75E-09 16149/14961/14960/14969/14972/15018/14964/13040/12526
Environmental InformationProcessing mmu04514 Cell adhesion molecules 10 174 5.39E-10 3.29E-08 14961/14960/14969/19264/14972/15018/14964/12741/80883/12526
Human Diseases mmu05330 Allograft rejection 6 63 1.12E-07 3.42E-06 14961/14960/14969/14972/15018/14964
Human Diseases mmu05332 Graft-versus-host disease 6 63 1.12E-07 3.42E-06 14961/14960/14969/14972/15018/14964
Human Diseases mmu04940 Type I diabetes mellitus 6 70 2.13E-07 5.19E-06 14961/14960/14969/14972/15018/14964
Cellular Processes mmu04145 Phagosome 8 182 2.97E-07 6.05E-06 14961/14960/14969/17533/14972/15018/14964/13040
Human Diseases mmu05320 Autoimmune thyroid disease 6 79 4.40E-07 7.66E-06 14961/14960/14969/14972/15018/14964
Human Diseases mmu05323 Rheumatoid arthritis 6 87 7.82E-07 1.13E-05 14961/14960/14969/14254/20315/20304
Human Diseases mmu05416 Viral myocarditis 6 88 8.37E-07 1.13E-05 14961/14960/14969/14972/15018/14964
Organismal Systems mmu04672 Intestinal immune network for IgA production 4 43 1.96E-05 0.000234487 14961/14960/14969/20315
Human Diseases mmu05169 Epstein-Barr virus infection 7 231 2.11E-05 0.000234487 14961/14960/14969/14972/15018/14964/12502
Organismal Systems mmu04640 Hematopoietic cell lineage 5 94 2.55E-05 0.000259539 14961/14960/14969/12502/12526
Human Diseases mmu05166 Human T-cell leukemia virus 1 infection 7 247 3.26E-05 0.000305554 14961/14960/14969/14972/15018/14964/12502
Human Diseases mmu05152 Tuberculosis 6 180 5.23E-05 0.00045538 16149/14961/14960/14969/17533/13040
Human Diseases mmu05150 Staphylococcus aureus infection 5 124 9.65E-05 0.000784943 14961/14960/14969/12260/12259
Human Diseases mmu05310 Asthma 3 25 0.00010994 0.000838268 14961/14960/14969
Human Diseases mmu05322 Systemic lupus erythematosus 5 148 0.00022194 0.001592726 14961/14960/14969/12260/12259
Human Diseases mmu05168 Herpes simplex virus 1 infection 8 458 0.00025292 0.001714244 16149/14961/14960/14969/14972/15018/14964/20304
Organismal Systems mmu04658 Th1 and Th2 cell differentiation 4 88 0.00032733 0.002101812 14961/14960/14969/12502
Human Diseases mmu05163 Human cytomegalovirus infection 6 256 0.00036073 0.002200437 14972/15018/14964/18595/20315/20304
Human Diseases mmu05142 Chagas disease 4 103 0.00059608 0.003428519 12502/12260/20304/12259
Organismal Systems mmu04659 Th17 cell differentiation 4 104 0.00061826 0.003428519 14961/14960/14969/12502
Human Diseases mmu05321 Inflammatory bowel disease 3 62 0.00164183 0.00870883 14961/14960/14969
Human Diseases mmu05140 Leishmaniasis 3 70 0.0023275 0.011831473 14961/14960/14969
Human Diseases mmu05164 Influenza A 4 173 0.00402625 0.01958829 14961/14960/14969/20304
Organismal Systems mmu04970 Salivary secretion 3 86 0.00417455 0.01958829 17105/16531/98660
Organismal Systems mmu04610 Complement and coagulation cascades 3 93 0.00519817 0.023488022 22370/12260/12259
Organismal Systems mmu04660 T cell receptor signaling pathway 3 103 0.00690329 0.030078638 19264/12502/12526
Human Diseases mmu05340 Primary immunodeficiency 2 36 0.00819423 0.033644766 19264/12526
Human Diseases mmu05145 Toxoplasmosis 3 110 0.0082733 0.033644766 14961/14960/14969
Human Diseases mmu05170 Human immunodeficiency virus 1 infection 4 240 0.01258496 0.049527889 14972/15018/14964/12502
Human Diseases mmu05030 Cocaine addiction 2 48 0.01425945 0.054364133 13162/214084
Cellular Processes mmu04144 Endocytosis 4 273 0.01934939 0.071534113 14972/15018/14964/18595
Human Diseases mmu05031 Amphetamine addiction 2 69 0.02821946 0.10125806 13162/214084
Organismal Systems mmu04360 Axon guidance 3 181 0.0310338 0.108174956 20315/19649/80883
Cellular Processes mmu04218 Cellular senescence 3 184 0.0323616 0.109669863 14972/15018/14964
Organismal Systems mmu04721 Synaptic vesicle cycle 2 77 0.0345386 0.110887076 13162/214084
Human Diseases mmu05133 Pertussis 2 77 0.0345386 0.110887076 12260/12259
Cellular Processes mmu04510 Focal adhesion 3 201 0.04044437 0.126518269 14254/18595/22370
Organismal Systems mmu04911 Insulin secretion 2 86 0.04223985 0.128831531 16531/98660
Human Diseases mmu05165 Human papillomavirus infection 4 362 0.04744758 0.141185479 14972/15018/14964/22370
Environmental InformationProcessing mmu04061 Viral protein interaction with cytokine and cytokine receptor 2 95 0.05052205 0.146754511 20315/20304
Human Diseases mmu05167 Kaposi sarcoma-associated herpesvirus infection 3 225 0.05344206 0.147950092 14972/15018/14964
Organismal Systems mmu04976 Bile secretion 2 100 0.05535742 0.147950092 28250/98660
Metabolism mmu00604 Glycosphingolipid biosynthesis - ganglio series 1 15 0.05572295 0.147950092 15212
Human Diseases mmu05203 Viral carcinogenesis 3 229 0.05578446 0.147950092 14972/15018/14964
Metabolism mmu00603 Glycosphingolipid biosynthesis - globo and isoglobo series 1 16 0.05932868 0.154002092 15212
Organismal Systems mmu04974 Protein digestion and absorption 2 108 0.06341896 0.161189846 67760/98660
Metabolism mmu00511 Other glycan degradation 1 18 0.06650009 0.165571647 15212
Organismal Systems mmu04972 Pancreatic secretion 2 114 0.06971217 0.170097692 16531/98660
Organismal Systems mmu04670 Leukocyte transendothelial migration 2 118 0.07401858 0.177064045 20315/12741
Metabolism mmu00531 Glycosaminoglycan degradation 1 21 0.07715781 0.177945404 15212
Organismal Systems mmu04650 Natural killer cell mediated cytotoxicity 2 121 0.07730415 0.177945404 14972/14964
Organismal Systems mmu04964 Proximal tubule bicarbonate reclamation 1 22 0.08068406 0.179954098 98660
Human Diseases mmu05020 Prion disease 3 268 0.08112685 0.179954098 12260/20304/12259
Cellular Processes mmu04142 Lysosome 2 131 0.08858196 0.192982118 15212/13040
Organismal Systems mmu04728 Dopaminergic synapse 2 135 0.09322538 0.199535019 13162/214084
Organismal Systems mmu04960 Aldosterone-regulated sodium reabsorption 1 38 0.135357 0.283930181 98660
Environmental InformationProcessing mmu04022 cGMP-PKG signaling pathway 2 173 0.14045784 0.283930181 16531/98660
Metabolism mmu00513 Various types of N-glycan biosynthesis 1 40 0.14196509 0.283930181 15212
Cellular Processes mmu04216 Ferroptosis 1 40 0.14196509 0.283930181 26570
Environmental InformationProcessing mmu04151 PI3K-Akt signaling pathway 3 359 0.15547602 0.305936688 14254/18595/22370
Organismal Systems mmu04062 Chemokine signaling pathway 2 192 0.1657251 0.320079762 20315/20304
Organismal Systems mmu04973 Carbohydrate digestion and absorption 1 48 0.1679107 0.320079762 98660
Metabolism mmu00520 Amino sugar and nucleotide sugar metabolism 1 50 0.17427715 0.327104803 15212
Human Diseases mmu05034 Alcoholism 2 205 0.18346392 0.334068629 13162/214084
Human Diseases mmu05205 Proteoglycans in cancer 2 205 0.18346392 0.334068629 13179/22370
Organismal Systems mmu04978 Mineral absorption 1 54 0.18686853 0.335264132 98660
Environmental InformationProcessing mmu04015 Rap1 signaling pathway 2 214 0.19591401 0.346398681 14254/18595
Cellular Processes mmu04810 Regulation of actin cytoskeleton 2 220 0.20427806 0.356027482 18595/20315
Organismal Systems mmu04961 Endocrine and other factor-regulated calcium reabsorption 1 61 0.20845644 0.358192758 98660
Organismal Systems mmu04623 Cytosolic DNA-sensing pathway 1 63 0.21452155 0.363494841 20304
Environmental InformationProcessing mmu04014 Ras signaling pathway 2 232 0.2211316 0.369562391 14254/18595
Environmental InformationProcessing mmu04020 Calcium signaling pathway 2 240 0.23244122 0.376205508 14254/18595
Human Diseases mmu05230 Central carbon metabolism in cancer 1 69 0.23244746 0.376205508 18595
Human Diseases mmu05218 Melanoma 1 72 0.24126088 0.376205508 18595
Human Diseases mmu05012 Parkinson disease 2 247 0.24237343 0.376205508 13162/214084
Human Diseases mmu05171 Coronavirus disease - COVID-19 2 247 0.24237343 0.376205508 12260/12259
Organismal Systems mmu04918 Thyroid hormone synthesis 1 74 0.24708184 0.376205508 98660
Human Diseases mmu05214 Glioma 1 74 0.24708184 0.376205508 18595
Organismal Systems mmu04971 Gastric acid secretion 1 75 0.24997604 0.376205508 98660
Organismal Systems mmu04924 Renin secretion 1 76 0.25285944 0.376205508 16531
Human Diseases mmu01521 EGFR tyrosine kinase inhibitor resistance 1 79 0.26144524 0.384293008 18595
Metabolism mmu00590 Arachidonic acid metabolism 1 86 0.28110804 0.3966762 19215
Cellular Processes mmu04540 Gap junction 1 86 0.28110804 0.3966762 18595
Organismal Systems mmu04260 Cardiac muscle contraction 1 87 0.2838752 0.3966762 98660
Environmental InformationProcessing mmu04512 ECM-receptor interaction 1 88 0.28663202 0.3966762 22370
Human Diseases mmu05235 PD-L1 expression and PD-1 checkpoint pathway in cancer 1 88 0.28663202 0.3966762 12502
Organismal Systems mmu04727 GABAergic synapse 1 89 0.28937854 0.3966762 67760
Organismal Systems mmu04666 Fc gamma R-mediated phagocytosis 1 92 0.29755666 0.403354582 19264
Environmental InformationProcessing mmu04350 TGF-beta signaling pathway 1 95 0.3056434 0.405295332 13179
Environmental InformationProcessing mmu04060 Cytokine-cytokine receptor interaction 2 292 0.30649575 0.405295332 20315/20304
Environmental InformationProcessing mmu04010 MAPK signaling pathway 2 294 0.30933918 0.405295332 14254/18595
Human Diseases mmu05231 Choline metabolism in cancer 1 98 0.31363975 0.405295332 18595
Human Diseases mmu05215 Prostate cancer 1 99 0.31628528 0.405295332 18595
Organismal Systems mmu04620 Toll-like receptor signaling pathway 1 100 0.31892092 0.405295332 20304
Organismal Systems mmu04925 Aldosterone synthesis and secretion 1 102 0.32416264 0.407709707 98660
Environmental InformationProcessing mmu04064 NF-kappa B signaling pathway 1 105 0.33195187 0.413246206 20315
Environmental InformationProcessing mmu04668 TNF signaling pathway 1 113 0.3522996 0.42857075 20304
Organismal Systems mmu04724 Glutamatergic synapse 1 113 0.3522996 0.42857075 67760
Environmental InformationProcessing mmu04066 HIF-1 signaling pathway 1 114 0.35480038 0.42857075 14254
Organismal Systems mmu04919 Thyroid hormone signaling pathway 1 120 0.36960938 0.442081809 98660
Organismal Systems mmu04726 Serotonergic synapse 1 131 0.39590696 0.46893834 214084
Human Diseases mmu05135 Yersinia infection 1 134 0.40289221 0.472623551 12526
Cellular Processes mmu04210 Apoptosis 1 136 0.40750542 0.473482487 13040
Organismal Systems mmu04270 Vascular smooth muscle contraction 1 143 0.42338082 0.487287362 16531
Human Diseases mmu05162 Measles 1 146 0.43005747 0.490345899 12502
Environmental InformationProcessing mmu04072 Phospholipase D signaling pathway 1 149 0.43665905 0.493263005 18595
Organismal Systems mmu04261 Adrenergic signaling in cardiomyocytes 1 152 0.4431864 0.49604349 98660
Human Diseases mmu05160 Hepatitis C 1 165 0.47063323 0.51934756 12741
Cellular Processes mmu04530 Tight junction 1 167 0.47473753 0.51934756 12741
Environmental InformationProcessing mmu04630 JAK-STAT signaling pathway 1 168 0.47677809 0.51934756 18595
Organismal Systems mmu04621 NOD-like receptor signaling pathway 1 211 0.55760202 0.600636142 20304
Environmental InformationProcessing mmu04024 cAMP signaling pathway 1 215 0.56447213 0.600636142 98660
Human Diseases mmu05417 Lipid and atherosclerosis 1 216 0.56617341 0.600636142 20304
Human Diseases mmu05202 Transcriptional misregulation in cancer 1 223 0.57790308 0.607794613 14254
Human Diseases mmu05200 Pathways in cancer 2 543 0.62189255 0.645958812 18595/20315
Human Diseases mmu05132 Salmonella infection 1 253 0.62477984 0.645958812 19264
Human Diseases mmu05206 MicroRNAs in cancer 1 303 0.69190774 0.709350794 18595
Environmental InformationProcessing mmu04080 Neuroactive ligand-receptor interaction 1 358 0.75229312 0.764831336 21334
Human Diseases mmu05022 Pathways of neurodegeneration - multiple diseases 1 472 0.84310653 0.850074348 13162
NA mmu01100 Metabolic pathways 2 1573 0.9888085 0.988808495 19215/15212

CD74 expression is elevated in Nurr1 knockdown mice

To ascertain whether CD74 expression is altered in Nurr1 knockdown mouse cells, we performed double immunofluorescence staining. This experiment was performed as part of the second phase of the study in which Nurr1 LV_shRNA was injected into the left striatum of the mouse brain, while the right striatum was injected with Nurr1 LV_Scramble as a control (n = 10). Dual immunofluorescence analysis indicated that the elevated CD74 expression observed in the Nurr1 knockdown group relative to the control group corresponds to dopaminergic neuron degeneration, as indicated by a decrease in TH and Nurr1 immunoreactive neurons (Figure 8).

Figure 8.

Figure 8

Dual immunofluorescence analysis of CD74 expression in the substantia nigra of a Nurr1 knockdown mouse model (n = 5).

CD74 (red, Alexa Fluor 594) is expressed at noticeably higher levels in the Nurr1 knockdown side of the brain compared to the control side. Dual immunofluorescence analysis indicated that the elevated CD74 expression observed in the Nurr1 knockdown group relative to the control group corresponds to the degeneration of dopaminergic neurons, as indicated by decreased numbers of TH (green, Alexa Fluor 488) and NURR1 (green, Alexa Fluor 488) immunoreactive neurons. Scale bars: 100 μm. CD74: Cluster of differentiation 74; DAPI: 4′,6-diamidino-2-phenylindole; Nurr1: nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; TH: tyrosine hydroxylase.

Cd74 expression is increased in a 6-OHDA-induced mouse model of PD

To confirm the above results in a PD mouse model, mouse brains were injected 6-OHDA for 4 weeks, after which the midbrain was removed and processed. Cd74 expression in the midbrain tissues was then assessed by qPCR. In comparison to the control side, which received injections of normal saline, there was a 1-fold increase in Cd74 mRNA expression in the tissues exposed to 6-OHDA (P = 0.0141, t = 2.871, df = 12; Figure 9).

Figure 9.

Figure 9

Elevated Cd74 mRNA expression in the substantia nigra of a 6-OHDA-induced model of Parkinson’s disease model as compared to the control side.

To generate the model, 6-OHDA and saline solution were injected into the left and right striatum, respectively. The data are presented as the mean ± SD (n = 7). *P < 0.05 (Student’s t-test). 6-OHDA: 6-Hydroxydopamine; Cd74: cluster of differentiation 74; SNc: substantia nigra compacta.

Discussion

A remarkable number of patients who have been diagnosed with PD exhibit decreased expression of the transcription factor Nurr1 (Chu et al., 2006; Liu et al., 2012; Li et al., 2018; Kambey et al., 2021a). The decrease in dopaminergic neurons manifested by diminished TH and dopamine transporter expression after Nurr1 knockdown is consistent with findings from other studies, including studies in humans (Le et al., 1999, 2008; Chu et al., 2002, 2006). However, how does Nurr1 knockdown affect dopaminergic neurons as well as non-neuronal cells? The first possibility is that Nurr1 is expressed in non-neuronal cell types and that Nurr1 mRNA is activated in macrophages by inflammatory stimuli such as lipopolysaccharide (Barish et al., 2005; Pei et al., 2005). Nurr1 is expressed by a substantial number of glial cells, including Müller cells, the most common type of glial cell in the retina (Kubrusly et al., 2008). Another possibility could involve exosomes. Exosomes are extracellular vesicles produced by cells on a regular basis, with events such as cellular stress, cell signaling, and injury, influencing their release (Mathivanan et al., 2010). Meng et al. (2020) observed transfer of α-synuclein (α-syn) from neurons to astrocytes via exosomes, with consequent detrimental effects on astrocytes. They first investigated if exosomes generated from methamphetamine (METH)-treated neurons contain abnormal α-syn. Exosomes produced by METH-treated cells were found to carry pathogenic α-syn in both animal and cell line coculture models. Moreover, α-syn accumulation and inflammatory reactions were induced in primary cultured astrocytes by the presence of METH-exosomes in the medium. METH or α-syn decreased Nurr1 expression and increased inflammatory cytokine levels in primary cultured astrocytes (Chen et al., 2020b; Meng et al., 2020). This may imply that the effects of Nurr1 knockdown could be transferred from neuronal cells to glial cells via exosomes.

The discovery of genes responsible for PD fostered optimism that they could be used to develop new therapies (Sardi et al., 2018). Unfortunately, the journey from defective gene to new treatment option is often challenging because not all genes code for proteins, and the function of the encoded protein is not always fully understood. One strategy for addressing these issues is to infer the functional impact of gene variants by evaluating the proteins encoded by certain PD-associated genes. Proteomics approaches are being used to comprehend disease mechanisms and uncover biomarkers of disease (Chapman et al., 2014; Bracht et al., 2015). When it comes to studying potential disease-associated proteins, proteomics-based techniques deliver results that are objective, high-throughput, and quantitative (Cristea et al., 2004; Savage, 2015). In our study, we found elevated levels of genes encoding proteins that are associated with the PD pathway. This is in concordance with previously published studies. As an example, a substrate-specific proteomics study of PD showed that parkin, an E3 ubiquitin ligase implicated in PD, targets outer mitochondrial membrane proteins (Li and Cookson, 2019). In more recent work, IP followed by label-free quantitative MS linked leucine-rich repeat kinase 2 to the wingless/integrated (WNT) signaling pathway (Salašová et al., 2017; Pellegrini et al., 2018). Similarly, proteomics analysis has provided new insights into Alzheimer’s disease etiology, allowing the identification of candidate biomarkers for future diagnostic and therapeutic approaches (Zhang et al., 2018).

Inflammatory responses are a significant component of all central nervous system (CNS) diseases, particularly neurodegenerative disorders. Using single-nuclei RNA technology, our discovery elucidated the previously unknown mechanism by which Nurr1 knockdown causes dopaminergic neuron death in PD. We identified several upregulated (activated) genes, the majority of which encode components of major histocompatibility complex II; the top 10 upregulated genes were Cd74, H2-Ab1, H2-Aa, H2-Eb1, lyz2, Mrc1, Slc6a3, Slc47a1, Ms4a4b, and Ptprc2. Unlike nonspecific neuroinflammatory agents like lipopolysaccharide (LPS), Nurr1 deficiency is mechanistically linked to PD. A recent study that combined single-cell RNA sequencing with multicolor flow cytometry to profile microglia in the brains of mice injected with LPS found distinct profiles of activated microglia under inflammatory conditions that differ greatly from the proinflammatory profiles associated with neurodegenerative disease (Sousa et al., 2018). During PD progression, the brain’s resident macrophages become morphologically activated (Perry, 2012). Neuronal degeneration, whether induced by acute tissue injury or by a neurodegenerative condition that progresses over time, is correlated with activation of microglia, the resident macrophages in the brain. The microglia have been dubbed “pathology sensors” because they are extremely sensitive to all types of brain injury (Kreutzberg, 1996). When the brain is injured or affected by disease, microglia alter their morphology and begin to upregulate or express de novo a number of myeloid antigens (Perry, 2012). An important and intriguing aspect of our findings is that the highly activated genes were expressed in oligodendrocyte clusters. This is consistent with recent results from Errea and Rodriguez-Oroz (2021), lending validity to our findings. They published a human single-nuclei transcriptome atlas of the substantia nigra and discovered a strong link between PD susceptibility and oligodendrocyte-specific gene regulation, implying that oligodendrocytes may play a causative role in PD (Errea and Rodriguez-Oroz, 2021). The role of Cd74 in the brain ailments has recently been explored and verified using scRNA-seq techniques that compared healthy brains and brains compromised by autoimmune disorders, neurodegeneration, or ischemia (Gosselin et al., 2017; Jordão et al., 2019). Moreover, Cd74 upregulation was recently observed in macrophages in the context of disease (Jordão et al., 2019). Our results regarding Cd74 expression in the Nurr1 knockdown model and an increase in Cd74 mRNA expression in a 6-OHDA-induced PD mouse model warrant further investigation.

To justify the relevance of major histocompatibility II-related genes in PD and other neurodegenerative disorders, Harms et al. (2013) used an in vivo mouse model produced by viral overexpression of α-syn as well as in vitro systems to investigate the role of the MHCII complex in α-syn-driven neuroinflammation and neurodegeneration. In an α-syn-induced rat model of PD, T cell infiltration and MHCII overexpression in microglia lead to accelerated neuronal degeneration (Subbarayan et al., 2020). Genetic studies have linked SNPs in the HLA-DR gene to sporadic late-onset PD (Hamza et al., 2010). Myriad studies have reported a link between PD and single-nucleotide polymorphisms in the MHC class II gene (Wissemann et al., 2013; Zhu et al., 2015; Chang et al., 2020), and mutation of the MHC class II gene results in similar symptoms to Alzheimer’s disease (Mattiace et al., 1990; Perlmutter et al., 1992). These studies suggest that the MCH II pathway is critical to neuroinflammation in PD.

A limitation of this study is that we did not target specific glial cells when exploring the mechanism by which Nurr1 knockdown affects dopaminergic neurons. Nurr1 affects dopaminergic neurons through glial cells (microglia, astrocytes, oligodendrocytes) (Jakaria et al., 2019; Oh et al., 2020). The use of cre and lox mice targeting specific cells, for example Nurr1Cd11b-cre (deletion of Nurr1 in microglia), Nurr1Oligo2-cre (deletion of Nurr1 in oligodendrocytes), or Nurr1Gfap-cre (deletion of Nurr1 in astrocytes), would have probably provided a different picture of the effect of Nurr1 deficiency on dopaminergic neurons. In conclusion, our findings demonstrate the mechanism for Nurr1-deficiency-based disruption of dopaminergic neurons in PD and provide a potential target for future drug development.

Additional files:

Additional file 1 (735.7KB, pdf) : Detailed in vitro, quantitative polymerase chain reaction, and immunohistochemistry experiments.

Additional file 1

Detailed in vitro, qu antitative polymerase chain reaction, and immunohistochemistry experiments

NRR-18-2037_Suppl1.pdf (735.7KB, pdf)

Additional file 2 (93.9KB, pdf) : Open peer review report 1.

Additional file 2
NRR-18-2037_Suppl2.pdf (93.9KB, pdf)

Additional Figure 1 (77.9KB, pdf) : The mechanism on how the shRNA plasmid gene silencers were produced.

Additional Figure 1

The mechanism on how the shRNA plasmid gene silencers were produced.

shRNA: short hairpin RNA; siRNA: silencer RNA.

NRR-18-2037_Suppl3.pdf (77.9KB, pdf)

Additional Figure 2 (118.1KB, pdf) : The gene ontology of mass spectrometry (LC-MS/MS) results.

Additional Figure 2

The gene ontology of mass spectrometry (LC-MS/MS) results.

LC-MS/MS: Liquid chromatography coupled with tandem mass spectrometry.

NRR-18-2037_Suppl4.pdf (118.1KB, pdf)

Additional Figure 3 (97.8KB, pdf) : Distribution of basic cell information in each sample before and after cell filtration.

Additional Figure 3

Distribution of basic cell information in each sample before and after cell filtration.

nCount_RNA: Total number of molecules detected; nFeature_RNA: comparable number of unique molecular identifiers; Nurr1: nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; Percent.mt: mitochondrial mRNA percentages.

NRR-18-2037_Suppl5.pdf (97.8KB, pdf)

Additional Table 1 (376.4KB, pdf) : Differentially expressed proteins on the label-free liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS).

Additional Table 1

Differentially expressed proteins on the label-free liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS).

NRR-18-2037_Suppl6.pdf (376.4KB, pdf)

Additional Table 2 (4MB, pdf) : “FindAllMarkers” genes enrichment.

Additional Table 2

“FindAllMarkers” genes enrichment.

Additional Table 3: Pathway's annotation.

Additional Table 4 (1.9MB, pdf) : The Gene Ontology biological process annotation function of all marker genes.

Additional Table 4

The Gene Ontology biological process annotation function of all marker genes.

NRR-18-2037_Suppl8.pdf (1.9MB, pdf)

Footnotes

Author contributions: In liaison with DSG, PAK serves as the investigative leader responsible for the development of study concepts and designs, experimental work, data analysis, and paper writing. WYL and JW contributed to data collection and analysis, BB contributed to the bio-informatics analysis. IQ and KK contributed to the write-up and proofread the final draft. The final manuscript was read and approved by all authors.

Conflicts of interest: The authors assert that they do not have any competing interests.

Data availability statement: All relevant data are within the paper and its Additional files.

Open peer reviewer: Sergiu Groppa, University of Mainz, Germany.

C-Editor: Zhao M; S-Editors: Yu J, Li CH; L-Editors: Crow E, Song LP; T-Editor: Jia Y

References

  • 1.Abdelgawad ME, Desterke C, Uzan G, Naserian S. Single-cell transcriptomic profiling and characterization of endothelial progenitor cells:new approach for finding novel markers. Stem Cell Res Ther. 2021;12:145. doi: 10.1186/s13287-021-02185-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Aoi M, Date I, Tomita S, Ohmoto T. The effect of intrastriatal single injection of GDNF on the nigrostriatal dopaminergic system in hemiparkinsonian rats:behavioral and histological studies using two different dosages. Neurosci Res. 2000;36:319–325. doi: 10.1016/s0168-0102(00)00097-3. [DOI] [PubMed] [Google Scholar]
  • 3.Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, Chak S, Naikawadi RP, Wolters PJ, Abate AR, Butte AJ, Bhattacharya M. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20:163–172. doi: 10.1038/s41590-018-0276-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Aranguren ME, Bechhofer S, Lord P, Sattler U, Stevens R. Understanding and using the meaning of statements in a bio-ontology:recasting the Gene Ontology in OWL. BMC Bioinformatics. 2007;8:57. doi: 10.1186/1471-2105-8-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Balu DT, Hoshaw BA, Malberg JE, Rosenzweig-Lipson S, Schechter LE, Lucki I. Differential regulation of central BDNF protein levels by antidepressant and non-antidepressant drug treatments. Brain Res. 2008;1211:37–43. doi: 10.1016/j.brainres.2008.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Barish GD, Downes M, Alaynick WA, Yu RT, Ocampo CB, Bookout AL, Mangelsdorf DJ, Evans RM. A nuclear receptor atlas:macrophage activation. Mol Endocrinol. 2005;19:2466–2477. doi: 10.1210/me.2004-0529. [DOI] [PubMed] [Google Scholar]
  • 7.Booeshaghi AS, Pachter L. Normalization of single-cell RNA-seq counts by log(x +1)*or log(1 +x) Bioinformatics. 2021;37:2223–2224. doi: 10.1093/bioinformatics/btab085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Borlongan CV, Sanberg PR. Elevated body swing test:a new behavioral parameter for rats with 6-hydroxydopamine-induced hemiparkinsonism. J Neurosci. 1995;15:5372–5378. doi: 10.1523/JNEUROSCI.15-07-05372.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bracht T, Schweinsberg V, Trippler M, Kohl M, Ahrens M, Padden J, Naboulsi W, Barkovits K, Megger DA, Eisenacher M, Borchers CH, Schlaak JF, Hoffmann AC, Weber F, Baba HA, Meyer HE, Sitek B. Analysis of disease-associated protein expression using quantitative proteomics—fibulin-5 is expressed in association with hepatic fibrosis. J Proteome Res. 2015;14:2278–2286. doi: 10.1021/acs.jproteome.5b00053. [DOI] [PubMed] [Google Scholar]
  • 10.Can A, Dao DT, Terrillion CE, Piantadosi SC, Bhat S, Gould TD. The tail suspension test. J Vis Exp. 2012:e3769. doi: 10.3791/3769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chang CF, Lin SZ, Chiang YH, Morales M, Chou J, Lein P, Chen HL, Hoffer BJ, Wang Y. Intravenous administration of bone morphogenetic protein-7 after ischemia improves motor function in stroke rats. Stroke. 2003;34:558–564. doi: 10.1161/01.str.0000051507.64423.00. [DOI] [PubMed] [Google Scholar]
  • 12.Chang KH, Wu YR, Chen YC, Fung HC, Chen CM. Association of genetic variants within HLA-DR region with Parkinson's disease in Taiwan. Neurobiol Aging. 2020;87:140.e13–140.e18. doi: 10.1016/j.neurobiolaging.2019.11.002. [DOI] [PubMed] [Google Scholar]
  • 13.Chapman JD, Goodlett DR, Masselon CD. Multiplexed and data-independent tandem mass spectrometry for global proteome profiling. Mass Spectrom Rev. 2014;33:452–470. doi: 10.1002/mas.21400. [DOI] [PubMed] [Google Scholar]
  • 14.Chen B, Garmire L, Calvisi DF, Chua MS, Kelley RK, Chen X. Harnessing big 'omics'data and AI for drug discovery in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2020a;17:238–251. doi: 10.1038/s41575-019-0240-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen L, Fan F, Wu L, Zhao Y. The nuclear receptor 4A family members:mediators in human disease and autophagy. Cell Mol Biol Lett. 2020b;25:48. doi: 10.1186/s11658-020-00241-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chen Z, Zhou L, Liu L, Hou Y, Xiong M, Yang Y, Hu J, Chen K. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat Commun. 2020c;11:5077. doi: 10.1038/s41467-020-18916-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chu Y, Kompoliti K, Cochran EJ, Mufson EJ, Kordower JH. Age-related decreases in Nurr1 immunoreactivity in the human substantia nigra. J Comp Neurol. 2002;450:203–214. doi: 10.1002/cne.10261. [DOI] [PubMed] [Google Scholar]
  • 18.Chu Y, Le W, Kompoliti K, Jankovic J, Mufson EJ, Kordower JH. Nurr1 in Parkinson's disease and related disorders. J Comp Neurol. 2006;494:495–514. doi: 10.1002/cne.20828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Clarke ZA, Andrews TS, Atif J, Pouyabahar D, Innes BT, MacParland SA, Bader GD. Tutorial:guidelines for annotating single-cell transcriptomic maps using automated and manual methods. Nat Protoc. 2021;16:2749–2764. doi: 10.1038/s41596-021-00534-0. [DOI] [PubMed] [Google Scholar]
  • 20.Craft RM, Mogil JS, Aloisi AM. Sex differences in pain and analgesia:the role of gonadal hormones. Eur J Pain. 2004;8:397–411. doi: 10.1016/j.ejpain.2004.01.003. [DOI] [PubMed] [Google Scholar]
  • 21.Cristea IM, Gaskell SJ, Whetton AD. Proteomics techniques and their application to hematology. Blood. 2004;103:3624–3634. doi: 10.1182/blood-2003-09-3295. [DOI] [PubMed] [Google Scholar]
  • 22.Decressac M, Volakakis N, Björklund A, Perlmann T. NURR1 in Parkinson disease--from pathogenesis to therapeutic potential. Nat Rev Neurol. 2013;9:629–636. doi: 10.1038/nrneurol.2013.209. [DOI] [PubMed] [Google Scholar]
  • 23.Decressac M, Kadkhodaei B, Mattsson B, Laguna A, Perlmann T, Björklund A. α-Synuclein-induced down-regulation of Nurr1 disrupts GDNF signaling in nigral dopamine neurons. Sci Transl Med. 2012;4:163ra156. doi: 10.1126/scitranslmed.3004676. [DOI] [PubMed] [Google Scholar]
  • 24.Dong J, Li S, Mo JL, Cai HB, Le WD. Nurr1-based therapies for Parkinson's disease. CNS Neurosci Ther. 2016;22:351–359. doi: 10.1111/cns.12536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Earls RH, Menees KB, Chung J, Barber J, Gutekunst CA, Hazim MG, Lee JK. Intrastriatal injection of preformed alpha-synuclein fibrils alters central and peripheral immune cell profiles in non-transgenic mice. J Neuroinflammation. 2019;16:250. doi: 10.1186/s12974-019-1636-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Errea O, Rodriguez-Oroz MC. Oligodendrocytes, a new player in the etiology of Parkinson's disease. Mov Disord. 2021;36:83. doi: 10.1002/mds.28393. [DOI] [PubMed] [Google Scholar]
  • 27.Fillingim RB, Ness TJ. Sex-related hormonal influences on pain and analgesic responses. Neurosci Biobehav Rev. 2000;24:485–501. doi: 10.1016/s0149-7634(00)00017-8. [DOI] [PubMed] [Google Scholar]
  • 28.Franzén O, Gan LM, Björkegren JLM. PanglaoDB:a web server for exploration of mouse and human single-cell RNA sequencing data. Database (Oxford) 20192019 doi: 10.1093/database/baz046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Galleguillos D, Fuentealba JA, Gómez LM, Saver M, Gómez A, Nash K, Burger C, Gysling K, Andrés ME. Nurr1 regulates RET expression in dopamine neurons of adult rat midbrain. J Neurochem. 2010;114:1158–1167. doi: 10.1111/j.1471-4159.2010.06841.x. [DOI] [PubMed] [Google Scholar]
  • 30.Gong L, Kwong DL, Dai W, Wu P, Li S, Yan Q, Zhang Y, Zhang B, Fang X, Liu L, Luo M, Liu B, Chow LK, Chen Q, Huang J, Lee VH, Lam KO, Lo AW, Chen Z, Wang Y, et al. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat Commun. 2021;12:1540. doi: 10.1038/s41467-021-21795-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O'Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK. An environment-dependent transcriptional network specifies human microglia identity. Science. 2017;356:eaal3222. doi: 10.1126/science.aal3222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Grün D, Muraro MJ, Boisset JC, Wiebrands K, Lyubimova A, Dharmadhikari G, van den Born M, van Es J, Jansen E, Clevers H, de Koning EJP, van Oudenaarden A. De novo prediction of stem cell identity using single-cell transcriptome data. Cell Stem Cell. 2016;19:266–277. doi: 10.1016/j.stem.2016.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, Kusel VI, Collura R, Roberts J, Griffith A, Samii A, Scott WK, Nutt J, Factor SA, Payami H. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nat Genet. 2010;42:781–785. doi: 10.1038/ng.642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hao Y, Hao S, Andersen-Nissen E, Mauck WM, 3rd, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, Hoffman P, Stoeckius M, Papalexi E, Mimitou EP, Jain J, Srivastava A, Stuart T, Fleming LM, Yeung B, Rogers AJ, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–3587.e29. doi: 10.1016/j.cell.2021.04.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Henderson JM, Annett LE, Ryan LJ, Chiang W, Hidaka S, Torres EM, Dunnett SB. Subthalamic nucleus lesions induce deficits as well as benefits in the hemiparkinsonian rat. Eur J Neurosci. 1999;11:2749–2757. doi: 10.1046/j.1460-9568.1999.00692.x. [DOI] [PubMed] [Google Scholar]
  • 36.Jakaria M, Haque ME, Cho DY, Azam S, Kim IS, Choi DK. Molecular insights into NR4A2(Nurr1):an emerging target for neuroprotective therapy against neuroinflammation and neuronal cell death. Mol Neurobiol. 2019;56:5799–5814. doi: 10.1007/s12035-019-1487-4. [DOI] [PubMed] [Google Scholar]
  • 37.Jankovic J, Chen S, Le WD. The role of Nurr1 in the development of dopaminergic neurons and Parkinson's disease. Prog Neurobiol. 2005;77:128–138. doi: 10.1016/j.pneurobio.2005.09.001. [DOI] [PubMed] [Google Scholar]
  • 38.Jordão MJC, Sankowski R, Brendecke SM, Sagar, Locatelli G, Tai YH, Tay TL, Schramm E, Armbruster S, Hagemeyer N, Groß O, Mai D, Çiçek Ö, Falk T, Kerschensteiner M, Grün D, Prinz M. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science. 2019;363:eaat7554. doi: 10.1126/science.aat7554. [DOI] [PubMed] [Google Scholar]
  • 39.Kaboord B, Perr M. Isolation of proteins and protein complexes by immunoprecipitation. Methods Mol Biol. 2008;424:349–364. doi: 10.1007/978-1-60327-064-9_27. [DOI] [PubMed] [Google Scholar]
  • 40.Kadkhodaei B, Alvarsson A, Schintu N, Ramsköld D, Volakakis N, Joodmardi E, Yoshitake T, Kehr J, Decressac M, Björklund A, Sandberg R, Svenningsson P, Perlmann T. Transcription factor Nurr1 maintains fiber integrity and nuclear-encoded mitochondrial gene expression in dopamine neurons. Proc Natl Acad Sci U S A. 2013;110:2360–2365. doi: 10.1073/pnas.1221077110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kadkhodaei B, Ito T, Joodmardi E, Mattsson B, Rouillard C, Carta M, Muramatsu S, Sumi-Ichinose C, Nomura T, Metzger D, Chambon P, Lindqvist E, Larsson NG, Olson L, Björklund A, Ichinose H, Perlmann T. Nurr1 is required for maintenance of maturing and adult midbrain dopamine neurons. J Neurosci. 2009;29:15923–15932. doi: 10.1523/JNEUROSCI.3910-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kambey PA, Kanwore K, Ayanlaja AA, Nadeem I, Du Y, Buberwa W, Liu W, Gao D. Failure of glial cell-line derived neurotrophic factor (GDNF) in clinical trials orchestrated by reduced NR4A2 (NURR1) transcription factor in Parkinson's disease. A systematic review. Front Aging Neurosci. 2021a;13:645583. doi: 10.3389/fnagi.2021.645583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kambey PA, Chengcheng M, Xiaoxiao G, Abdulrahman AA, Kanwore K, Nadeem I, Jiao W, Gao D. The orphan nuclear receptor Nurr1 agonist amodiaquine mediates neuroprotective effects in 6-OHDA Parkinson's disease animal model by enhancing the phosphorylation of P38 mitogen-activated kinase but not PI3K/AKT signaling pathway. Metab Brain Dis. 2021b;36:609–625. doi: 10.1007/s11011-021-00670-2. [DOI] [PubMed] [Google Scholar]
  • 44.Kim M, Min YK, Jang J, Park H, Lee S, Lee CH. Single-cell RNA sequencing reveals distinct cellular factors for response to immunotherapy targeting CD73 and PD-1 in colorectal cancer. J Immunother Cancer. 2021;9:e002503. doi: 10.1136/jitc-2021-002503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kreutzberg GW. Microglia:a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–318. doi: 10.1016/0166-2236(96)10049-7. [DOI] [PubMed] [Google Scholar]
  • 46.Kubrusly RC, Panizzutti R, Gardino PF, Stutz B, Reis RA, Ventura AL, de Mello MC, de Mello FG. Expression of functional dopaminergic phenotype in purified cultured Müller cells from vertebrate retina. Neurochem Int. 2008;53:63–70. doi: 10.1016/j.neuint.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 47.La Manno G, Gyllborg D, Codeluppi S, Nishimura K, Salto C, Zeisel A, Borm LE, Stott SRW, Toledo EM, Villaescusa JC, Lönnerberg P, Ryge J, Barker RA, Arenas E, Linnarsson S. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell. 2016;167:566–580.e19. doi: 10.1016/j.cell.2016.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Le W, Conneely OM, He Y, Jankovic J, Appel SH. Reduced Nurr1 expression increases the vulnerability of mesencephalic dopamine neurons to MPTP-induced injury. J Neurochem. 1999;73:2218–2221. [PubMed] [Google Scholar]
  • 49.Le W, Pan T, Huang M, Xu P, Xie W, Zhu W, Zhang X, Deng H, Jankovic J. Decreased NURR1 gene expression in patients with Parkinson's disease. J Neurol Sci. 2008;273:29–33. doi: 10.1016/j.jns.2008.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Le WD, Xu P, Jankovic J, Jiang H, Appel SH, Smith RG, Vassilatis DK. Mutations in NR4A2 associated with familial Parkinson disease. Nat Genet. 2003;33:85–89. doi: 10.1038/ng1066. [DOI] [PubMed] [Google Scholar]
  • 51.Li T, Yang Z, Li S, Cheng C, Shen B, Le W. Alterations of NURR1 and cytokines in the peripheral blood mononuclear cells:combined biomarkers for Parkinson's disease. Front Aging Neurosci. 2018;10:392. doi: 10.3389/fnagi.2018.00392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Li Y, Cookson MR. Proteomics;applications in familial Parkinson's disease. J Neurochem. 2019;151:446–458. doi: 10.1111/jnc.14708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu H, Wei L, Tao Q, Deng H, Ming M, Xu P, Le W. Decreased NURR1 and PITX3 gene expression in Chinese patients with Parkinson's disease. Eur J Neurol. 2012;19:870–875. doi: 10.1111/j.1468-1331.2011.03644.x. [DOI] [PubMed] [Google Scholar]
  • 54.Lou X, Liao W. Association of Nurr1 gene mutations with Parkinson's disease in the Han population living in the Hubei province of China. Neural Regen Res. 2012;7:1791–1796. doi: 10.3969/j.issn.1673-5374.2012.23.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mabbott NA, Baillie JK, Brown H, Freeman TC, Hume DA. An expression atlas of human primary cells:inference of gene function from coexpression networks. BMC Genomics. 2013;14:632. doi: 10.1186/1471-2164-14-632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mathivanan S, Ji H, Simpson RJ. Exosomes:extracellular organelles important in intercellular communication. J Proteomics. 2010;73:1907–1920. doi: 10.1016/j.jprot.2010.06.006. [DOI] [PubMed] [Google Scholar]
  • 57.Mattiace LA, Davies P, Dickson DW. Detection of HLA-DR on microglia in the human brain is a function of both clinical and technical factors. Am J Pathol. 1990;136:1101–1114. [PMC free article] [PubMed] [Google Scholar]
  • 58.Maxwell MA, Muscat GE. The NR4A subgroup:immediate early response genes with pleiotropic physiological roles. Nuclear receptor signaling. 2006;4:e002. doi: 10.1621/nrs.04002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mendez I, Sanchez-Pernaute R, Cooper O, Viñuela A, Ferrari D, Björklund L, Dagher A, Isacson O. Cell type analysis of functional fetal dopamine cell suspension transplants in the striatum and substantia nigra of patients with Parkinson's disease. Brain. 2005;128:1498–1510. doi: 10.1093/brain/awh510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Meng Y, Ding J, Li C, Fan H, He Y, Qiu P. Transfer of pathological α-synuclein from neurons to astrocytes via exosomes causes inflammatory responses after METH exposure. Toxicol Lett. 2020;331:188–199. doi: 10.1016/j.toxlet.2020.06.016. [DOI] [PubMed] [Google Scholar]
  • 61.Mogil JS, Chanda ML. The case for the inclusion of female subjects in basic science studies of pain. Pain. 2005;117:1–5. doi: 10.1016/j.pain.2005.06.020. [DOI] [PubMed] [Google Scholar]
  • 62.Montarolo F, Perga S, Martire S, Navone DN, Marchet A, Leotta D, Bertolotto A. Altered NR4A subfamily gene expression level in peripheral blood of Parkinson's and Alzheimer's disease patients. Neurotox Res. 2016;30:338–344. doi: 10.1007/s12640-016-9626-4. [DOI] [PubMed] [Google Scholar]
  • 63.No authors listed. Mouse Brain Atlas. 2001. [Accessed June 2021]. http://labs.gaidi.ca/mouse-brain-atlas/?ml=1.5&ap=0.5&dv=3.5.
  • 64.Oh M, Kim SY, Gil JE, Byun JS, Cha DW, Ku B, Lee W, Kim WK, Oh KJ, Lee EW, Bae KH, Lee SC, Han BS. Nurr1 performs its anti-inflammatory function by regulating RasGRP1 expression in neuro-inflammation. Sci Rep. 2020;10:10755. doi: 10.1038/s41598-020-67549-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018;18:35–45. doi: 10.1038/nri.2017.76. [DOI] [PubMed] [Google Scholar]
  • 66.Pei L, Castrillo A, Chen M, Hoffmann A, Tontonoz P. Induction of NR4A orphan nuclear receptor expression in macrophages in response to inflammatory stimuli. J Biol Chem. 2005;280:29256–29262. doi: 10.1074/jbc.M502606200. [DOI] [PubMed] [Google Scholar]
  • 67.Pellegrini L, Hauser DN, Li Y, Mamais A, Beilina A, Kumaran R, Wetzel A, Nixon-Abell J, Heaton G, Rudenko I, Alkaslasi M, Ivanina N, Melrose HL, Cookson MR, Harvey K. Proteomic analysis reveals co-ordinated alterations in protein synthesis and degradation pathways in LRRK2 knockout mice. Hum Mol Genet. 2018;27:3257–3271. doi: 10.1093/hmg/ddy232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, Browne WJ, Clark A, Cuthill IC, Dirnagl U, Emerson M, Garner P, Holgate ST, Howells DW, Karp NA, Lazic SE, Lidster K, MacCallum CJ, Macleod M, Pearl EJ, et al. The ARRIVE guidelines 2.0:Updated guidelines for reporting animal research. PLoS Biol. 2020;18:e3000410. doi: 10.1371/journal.pbio.3000410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Perlmutter LS, Scott SA, Barrón E, Chui HC. MHC class II-positive microglia in human brain:association with Alzheimer lesions. J Neurosci Res. 1992;33:549–558. doi: 10.1002/jnr.490330407. [DOI] [PubMed] [Google Scholar]
  • 70.Perry VH. Innate inflammation in Parkinson's disease. Cold Spring Harb Perspect Med. 2012;2:a009373. doi: 10.1101/cshperspect.a009373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rodríguez-Traver E, Solís O, Díaz-Guerra E, Ortiz Ó, Vergaño-Vera E, Méndez-Gómez HR, García-Sanz P, Moratalla R, Vicario-Abejón C. Role of Nurr1 in the generation and differentiation of dopaminergic neurons from stem cells. Neurotox Res. 2016;30:14–31. doi: 10.1007/s12640-015-9586-0. [DOI] [PubMed] [Google Scholar]
  • 72.Ruiz-Sánchez E, Yescas P, Rodríguez-Violante M, Martínez-Rodríguez N, Díaz-López JN, Ochoa A, Valdes-Rojas SS, Magos-Rodríguez D, Rojas-Castañeda JC, Cervantes-Arriaga A, Canizales-Quinteros S, Rojas P. Association of polymorphisms and reduced expression levels of the NR4A2 gene with Parkinson's disease in a Mexican population. J Neurol Sci. 2017;379:58–63. doi: 10.1016/j.jns.2017.05.029. [DOI] [PubMed] [Google Scholar]
  • 73.Salašová A, Yokota C, Potěšil D, Zdráhal Z, Bryja V, Arenas E. A proteomic analysis of LRRK2 binding partners reveals interactions with multiple signaling components of the WNT/PCP pathway. Mol Neurodegener. 2017;12:54. doi: 10.1186/s13024-017-0193-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sardi SP, Cedarbaum JM, Brundin P. Targeted therapies for Parkinson's disease:from genetics to the clinic. Mov Disord. 2018;33:684–696. doi: 10.1002/mds.27414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33:495–502. doi: 10.1038/nbt.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Savage N. Proteomics:High-protein research. Nature. 2015;527:S6–7. doi: 10.1038/527S6a. [DOI] [PubMed] [Google Scholar]
  • 77.Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ:25 years of image analysis. Nat Methods. 2012;9:671–675. doi: 10.1038/nmeth.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Schwarting RK, Goldenberg R, Steiner H, Fornaguera J, Huston JP. A video image analyzing system for open-field behavior in the rat focusing on behavioral asymmetries. J Neurosci Methods. 1993;49:199–210. doi: 10.1016/0165-0270(93)90125-b. [DOI] [PubMed] [Google Scholar]
  • 79.Seth S, Mallik S, Bhadra T, Zhao Z. Dimensionality reduction and louvain agglomerative hierarchical clustering for cluster-specified frequent biomarker discovery in single-cell sequencing data. Front Genet. 2022;13:828479. doi: 10.3389/fgene.2022.828479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sousa C, Golebiewska A, Poovathingal SK, Kaoma T, Pires-Afonso Y, Martina S, Coowar D, Azuaje F, Skupin A, Balling R, Biber K, Niclou SP, Michelucci A. Single-cell transcriptomics reveals distinct inflammation-induced microglia signatures. EMBO Rep. 2018;19:e46171. doi: 10.15252/embr.201846171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Subbarayan MS, Hudson C, Moss LD, Nash KR, Bickford PC. T cell infiltration and upregulation of MHCII in microglia leads to accelerated neuronal loss in an α-synuclein rat model of Parkinson's disease. J Neuroinflammation. 2020;17:242. doi: 10.1186/s12974-020-01911-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Tanapat P. Neuronal cell markers. Mater Methods. 2013;3:196. [Google Scholar]
  • 83.Textor J, van der Zander B, Gilthorpe MS, Liskiewicz M, Ellison GT. Robust causal inference using directed acyclic graphs:the R package 'dagitty'. Int J Epidemiol. 2016;45:1887–1894. doi: 10.1093/ije/dyw341. [DOI] [PubMed] [Google Scholar]
  • 84.Tian YM, Chen X, Luo DZ, Zhang XH, Xue H, Zheng LF, Yang N, Wang XM, Zhu JX. Alteration of dopaminergic markers in gastrointestinal tract of different rodent models of Parkinson's disease. Neuroscience. 2008;153:634–644. doi: 10.1016/j.neuroscience.2008.02.033. [DOI] [PubMed] [Google Scholar]
  • 85.Tikhonova AN, Dolgalev I, Hu H, Sivaraj KK, Hoxha E, Cuesta-Domínguez Á, Pinho S, Akhmetzyanova I, Gao J, Witkowski M, Guillamot M, Gutkin MC, Zhang Y, Marier C, Diefenbach C, Kousteni S, Heguy A, Zhong H, Fooksman DR, Butler JM, et al. The bone marrow microenvironment at single-cell resolution. Nature. 2019;569:222–228. doi: 10.1038/s41586-019-1104-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.UniProt Consortium. UniProt:the universal protein knowledgebase in 2021. Nucleic Acids Res. 2021;49:D480–489. doi: 10.1093/nar/gkaa1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wang X, Zhuang W, Fu W, Wang X, Lv E, Li F, Zhou S, Rausch WD, Wang X. The lentiviral-mediated Nurr1 genetic engineering mesenchymal stem cells protect dopaminergic neurons in a rat model of Parkinson's disease. Am J Transl Res. 2018;10:1583–1599. [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang Y, Wang Q, Huang H, Huang W, Chen Y, McGarvey PB, Wu CH, Arighi CN. A crowdsourcing open platform for literature curation in UniProt. PLoS Biol. 2021;19:e3001464. doi: 10.1371/journal.pbio.3001464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wissemann WT, Hill-Burns EM, Zabetian CP, Factor SA, Patsopoulos N, Hoglund B, Holcomb C, Donahue RJ, Thomson G, Erlich H, Payami H. Association of Parkinson disease with structural and regulatory variants in the HLA region. Am J Hum Genet. 2013;93:984–993. doi: 10.1016/j.ajhg.2013.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wu H, Wang C, Wu S. Single-cell sequencing for drug discovery and drug development. Curr Top Med Chem. 2017;17:1769–1777. doi: 10.2174/1568026617666161116145358. [DOI] [PubMed] [Google Scholar]
  • 91.Xu J, Zhang P, Huang Y, Zhou Y, Hou Y, Bekris LM, Lathia J, Chiang CW, Li L, Pieper AA, Leverenz JB, Cummings J, Cheng F. Multimodal single-cell/nucleus RNA sequencing data analysis uncovers molecular networks between disease-associated microglia and astrocytes with implications for drug repurposing in Alzheimer's disease. Genome Res. 2021;31:1900–1912. doi: 10.1101/gr.272484.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yang Z, Li T, Li S, Wei M, Qi H, Shen B, Chang RC, Le W, Piao F. Altered expression levels of microRNA-132 and Nurr1 in peripheral blood of Parkinson's disease:potential disease biomarkers. ACS Chem Neurosci. 2019;10:2243–2249. doi: 10.1021/acschemneuro.8b00460. [DOI] [PubMed] [Google Scholar]
  • 93.Yu C, Chen W, Wang Z, Lou H. Comparative proteomic analysis of tomato (Solanum lycopersicum L.) shoots reveals crosstalk between strigolactone and auxin. Genomics. 2021;113:3163–3173. doi: 10.1016/j.ygeno.2021.07.009. [DOI] [PubMed] [Google Scholar]
  • 94.Zetterström RH, Solomin L, Jansson L, Hoffer BJ, Olson L, Perlmann T. Dopamine neuron agenesis in Nurr1-deficient mice. Science. 1997;276:248–250. doi: 10.1126/science.276.5310.248. [DOI] [PubMed] [Google Scholar]
  • 95.Zhang C, Li H, Han R. An open-source video tracking system for mouse locomotor activity analysis. BMC Res Notes. 2020;13:48. doi: 10.1186/s13104-020-4916-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhang JS, Hou PP, Shao S, Manaenko A, Xiao ZP, Chen Y, Zhao B, Jia F, Zhang XH, Mei QY, Hu Q. microRNA-455-5p alleviates neuroinflammation in cerebral ischemia/reperfusion injury. Neural Regen Res. 2022;17:1769–1775. doi: 10.4103/1673-5374.332154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zhang Q, Ma C, Gearing M, Wang PG, Chin LS, Li L. Integrated proteomics and network analysis identifies protein hubs and network alterations in Alzheimer's disease. Acta Neuropathol Commun. 2018;6:19. doi: 10.1186/s40478-018-0524-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhang X, Lan Y, Xu J, Quan F, Zhao E, Deng C, Luo T, Xu L, Liao G, Yan M, Ping Y, Li F, Shi A, Bai J, Zhao T, Li X, Xiao Y. CellMarker:a manually curated resource of cell markers in human and mouse. Nucleic Acids Res. 2019;47:D721–728. doi: 10.1093/nar/gky900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Zhou HY, Huai YP, Jin X, Yan P, Tang XJ, Wang JY, Shi N, Niu M, Meng ZX, Wang X. An enriched environment reduces hippocampal inflammatory response and improves cognitive function in a mouse model of stroke. Neural Regen Res. 2022;17:2497–2503. doi: 10.4103/1673-5374.338999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zhu RL, Lu XC, Tang LJ, Huang BS, Yu W, Li S, Li LX. Association between HLA rs3129882 polymorphism and Parkinson's disease:a meta-analysis. Eur Rev Med Pharmacol Sci. 2015;19:423–432. [PubMed] [Google Scholar]
  • 101.Zou Q, Lv Y, Gan Z, Liao S, Liang Z. Identification and validation of a malignant cell subset marker-based polygenic risk score in stomach adenocarcinoma through integrated analysis of bulk and single-cell RNA sequencing data. Front Cell Dev Biol. 2021;9:720649. doi: 10.3389/fcell.2021.720649. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Additional file 1

Detailed in vitro, qu antitative polymerase chain reaction, and immunohistochemistry experiments

NRR-18-2037_Suppl1.pdf (735.7KB, pdf)
Additional file 2
NRR-18-2037_Suppl2.pdf (93.9KB, pdf)
Additional Figure 1

The mechanism on how the shRNA plasmid gene silencers were produced.

shRNA: short hairpin RNA; siRNA: silencer RNA.

NRR-18-2037_Suppl3.pdf (77.9KB, pdf)
Additional Figure 2

The gene ontology of mass spectrometry (LC-MS/MS) results.

LC-MS/MS: Liquid chromatography coupled with tandem mass spectrometry.

NRR-18-2037_Suppl4.pdf (118.1KB, pdf)
Additional Figure 3

Distribution of basic cell information in each sample before and after cell filtration.

nCount_RNA: Total number of molecules detected; nFeature_RNA: comparable number of unique molecular identifiers; Nurr1: nuclear receptor-related factor 1; Nurr1 LV_Scramble: Nurr1 Lentiviral_Scramble; Nurr1 LV_shRNA: Nurr1 Lentiviral_short hairpin RNA; Percent.mt: mitochondrial mRNA percentages.

NRR-18-2037_Suppl5.pdf (97.8KB, pdf)
Additional Table 1

Differentially expressed proteins on the label-free liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS).

NRR-18-2037_Suppl6.pdf (376.4KB, pdf)
Additional Table 2

“FindAllMarkers” genes enrichment.

Additional Table 4

The Gene Ontology biological process annotation function of all marker genes.

NRR-18-2037_Suppl8.pdf (1.9MB, pdf)

Articles from Neural Regeneration Research are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES