Next Article in Journal
Diagnosis and Assessment of Dental Caries Using Novel Bioactive Caries Detecting Dye Solution
Next Article in Special Issue
Exploring the Interaction of Indole-3-Acetonitrile with Neuroblastoma Cells: Understanding the Connection with the Serotonin and Dopamine Pathways
Previous Article in Journal
Does the SARS-CoV-2 Spike Receptor-Binding Domain Hamper the Amyloid Transformation of Alpha-Synuclein after All?
Previous Article in Special Issue
Distinctive Neuroanatomic Regions Involved in Cocaine-Induced Behavioral Sensitization in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

AgRP Neuron-Specific Ablation Represses Appetite, Energy Intake, and Somatic Growth in Larval Zebrafish

1
Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 202, Taiwan
2
Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
3
Division of Hemato-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung City 204, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(2), 499; https://doi.org/10.3390/biomedicines11020499
Submission received: 30 November 2022 / Revised: 1 February 2023 / Accepted: 6 February 2023 / Published: 9 February 2023
(This article belongs to the Special Issue Neuropeptides, Dopamine and Their Interactions in Neuroscience)

Abstract

:
Neuronal circuits regulating appetite are dominated by arcuate nucleus neurons, which include appetite-promoting and -suppressing neurons that release the orexigenic neuropeptide agouti-related protein (AgRP) and anorexigenic neuropeptide pro-opiomelanocortin, respectively, to compete for melanocortin receptors to modulate feeding behavior. In this study, we expressed novel agrp promoters, including different lengths of the 5’ flanking regions of the agrp gene (4749 bp) in the zebrafish genome. We used the agrp promoter to derive the enhanced green fluorescent protein (EGFP)-nitroreductase (NTR) fusion protein, allowing expression of the green fluorescence signal in the AgRP neurons. Then, we treated the transgenic zebrafish AgRP4.7NTR (Tg [agrp-EGFP-NTR]) with metronidazole to ablate the AgRP neurons in the larvae stage and observed a decline in their appetite and growth. The expression of most orexigenic and growth hormone/insulin-like growth factor axis genes decreased, whereas that of several anorexigenic genes increased. Our findings demonstrate that AgRP is a critical regulator of neuronal signaling for zebrafish appetite and energy intake control. Thus, AgRP4.7NTR can be used as a drug-screening platform for therapeutic targets to treat human appetite disorders, including obesity. Furthermore, the unique agrp promoter we identified can be a powerful tool for research on AgRP neurons, especially AgRP neuron-mediated pathways in the hypothalamus, and appetite.

1. Introduction

The arcuate nucleus (ARC) is located in the hypothalamus of the brain. Previous studies have revealed that ARC is crucial for food intake and energy regulation. This includes the appetite-promoting agouti-related protein (AgRP) neurons and the appetite-suppressing pro-opiomelanocortin (POMC) neurons, which can affect neural signal transmission and alter feeding behavior by controlling downstream melanocortin receptors. In the melanocortin system, leptin binds to the leptin receptor, activating POMC neurons, while inhibiting AgRP neurons [1,2,3,4,5]. Leptin and insulin receptors are highly abundant in AgRP and POMC neurons. When these receptors are activated by leptin and insulin, they can trigger the appetite control pathway of cells in the ARC [6,7,8].
AgRP is a neuropeptide produced by AgRP neurons in the ARC of the central nervous system [9,10]. AgRP has been shown to enhance hunger, slow metabolism, and lower energy consumption [10,11]. Under normal conditions, as the number of adipocytes decreases, the production of insulin and leptin is inhibited [12,13], allowing AgRP neurons to be activated and produce AgRP and neuropeptide Y (NPY) to convey body hunger cues that boost appetite, slow the metabolic rate, and limit energy consumption [9,14]. Appetite is repressed by leptin [15] and insulin [16] and triggered by ghrelin [17,18]. AgRP can be used as a competitive antagonist to suppress α-melanocyte-stimulating hormone (α-MSH) and increase appetite by attaching to the melanocortin 4 receptor (MC4R) and preventing α-MSH from binding to MC4R [19,20,21,22,23].
Mice lacking in the agrp gene are slimmer and exhibit severe hypophagia [24,25]. Ablation of AgRP neurons via administration of diphtheria toxin directly induces the Fos proto-oncogene, AP-1 transcription factor subunit, and gliosis, leading to appetite suppression [26,27]. Similarly, ablation of AgRP1 neurons in larval zebrafish resulted in decreased food intake [28]. The zebrafish melanocortin system also influences somatic growth [29]; morpholino-based agrp knockdown leads to lower expression of the growth hormone 1 (gh1) gene and shorter body length [23], while transgenic overexpression of AgRP [30] or deletion of α-MSH [31] shows the opposite effects.
Most previous studies on AgRP neurons in zebrafish used the gene knock-in technique to insert the target sequence before the agrp transcription start point (such as the AgRP–ires–Cre system) [26,32,33] or a bacterial artificial chromosome library containing the 5’-flanking region of agrp [28,34,35,36] to generate transgenic lines. However, the approach described in the literature has several limitations for agrp-related research, including low transmission rates, limited flexibility of transgenic constructs, and lack of switchability of transgenic genes. In this study, we sought to identify a novel agrp promoter for use in zebrafish AgRP neurons.
This study employed a nitroreductase (NTR) cell ablation system in order to test the functionality of the cloned agrp promoters [26]. NTR can convert the prodrug metronidazole (MTZ) into a cell-autonomous DNA crosslinking agent, thereby initiating apoptosis in the target cell of interest [37,38]. We generated AgRP4.7NTR-transgenic zebrafish and obtained NTR/MTZ-ablated AgRP neurons in the hypothalamus of zebrafish. Since AgRP neuron-specific ablation represses appetite and energy intake, resulting in agrp and other orexigenic gene depression, AgRP4.7NTR is a novel model for pharmacological studies to investigate these functions and network reorganization as orexin input is lost. In addition, we identified a unique agrp promoter that can be used as a potent tool for evaluating AgRP neurons, facilitating the investigation of AgRP neuron-mediated pathways in the hypothalamus, which will be useful for appetite research in the future.

2. Materials and Methods

2.1. Generation and Maintenance of Transgenic Zebrafish

In this study, the following transgenic zebrafish lines were developed and used: (a) AgRP4.7NTR (Tg [agrp-enhanced green fluorescent protein [EGFP]-NTR]), which showed AgRP neuron-specific expression of EGFP-NTR fusion protein driven by the 4.7 kb agrp promoter; (b) POMCNTR [Tg (pomc-EGFP-NTR)], which showed POMC neuron-specific expression of EGFP-NTR fusion protein driven by the pomc promoter [39]; and (c) β-actNTR [Tg (β-act-EGFP-NTR)], which showed global EGFP-NTR fusion protein expression under the control of the β-actin promoter. All animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) guidelines (approval ID: 1110209).
Transgenic zebrafish were maintained in a controlled environment with a 14/10-h light–dark cycle at 28 °C. All procedures were conducted in accordance with the IACUC guidelines of the National Yang Ming Chiao Tung University, Taipei, Taiwan.

2.2. Promoter Cloning

The 4.7 kb agrp promoter used in this study comprised a 5′ flanking region (4749 bp) at the transcription start site of the agrp gene and a partial 5′ coding region with an in-frame 9 bp downstream of the agrp translational start site, amplified with Q5 High-Fidelity DNA Polymerase (NEB, Ipswich, MA, USA) and the following primers: 5′-end primer, 5′-GTCGACCTCGAGGCTCTGCAAGTGAAGCTTCAGGTC-3′; and 3′-end primer, 5′-CACTAGTCCCAGCATCATAATCACTCAGACTCA-3′. The amplified 4.7 kb agrp promoter was purified using a FavorPrep GEL/PCR Purification Mini Kit (FAVORGEN, Ping Tung, Taiwan), according to the manufacturer’s instructions.
For isolation of the 5′ truncated sequences of the 4.7 kb agrp promoter, the 2.7, 3.4, and 4 kb agrp promoters were generated via polymerase chain reaction (PCR) amplification using the 3′-end primer 5′-CACTAGTCCCAGCATCATAATCACTCAGACTCA-3′, along with primers specific to different regions of the 5′-end agrp promoter. The specific primers for amplification of 2.7, 3.4, and 4 kb agrp promoters were AgRP2.7 (5′-GTCGACCTCGAGAATGAGGGAAGTTGTGGATACCCG-3′), AgRP3.4 (5′-GTCGACCTCGAGCTCTCACCAGCCCCTCTAGATAAC-3′), and AgRP4.0 (5′-GTCGACCTCGAGGTGCCTTGACCATTTAAATGTGAC-3′), respectively. All PCR products were cloned into the pGEM-T Easy vector (Promega, Madison, WI, USA).

2.3. Transcription Factor-Binding Site Prediction

The potential transcription factor-binding site of the cloned zebrafish agrp promoter was predicted using Table Browser “https://genome.ucsc.edu/cgi-bin/hgTables” (accessed on 20 October 2022). The datasheet on this website is based on the JASPAR database (version 2022) “https://jaspar.genereg.net/” (accessed on 20 October 2022).

2.4. MTZ Treatment

MTZ was administered to the embryos and juvenile zebrafish in a 6-well plate and a 3 L tank, respectively, at a concentration of 10 mM (Sigma-Aldrich, Burlington, MA, USA), with a daily water change. The embryos were treated with MTZ beginning at 2 days post-fertilization (dpf) and ending at 5 dpf. However, for long-term treatment after 7 dpf, the treatment concentration is reduced to 1 mM.

2.5. Quantitative Reverse Transcription PCR (RT-qPCR)

Total RNA was extracted from zebrafish tissue using TRIzol reagent (Thermo Fisher Scientific, USA) and column-purified with EasyPure Total RNA Spin Kit Tissue (Bioman, New Taipei City, Taiwan), per the manufacturer’s instructions. Then, 100 ng of purified RNA was reverse-transcribed with a first-strand cDNA synthesis kit (K1691; Thermo Fisher Scientific). For real-time reverse transcriptase quantitative PCR (RT-qPCR), a One-Step Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) was used. Table S1 lists the genes and their corresponding primer sequences used in this study. All investigated gene mRNA expression levels were normalized to bactin1 levels.

2.6. Whole Mount In Situ Hybridization (WISH)

The gene-specific probes used in WISH were cloned by PCR into a pGEM-T Easy vector (Promega, Madison, WI, USA) using the primers listed in Table S2. Antisense probes were created in vitro using a DIG RNA Labelling kit (SP6/T7; Roche Applied Science, Penzberg, Germany). WISH was performed per the procedure described in our previous study [40].

2.7. Quantification of Food Intake

To quantify the food intake by zebrafish, the method described by Shimada et al. [41] was used after modifications. Fluorescently tagged paramecia used as the larval fish meal were produced with the lipophilic tracer 4-(4-(didecylamino) styryl)-N-methylpyridinium iodide (4-Di-10-ASP; Invitrogen, Carlsbad, CA, USA). To facilitate free swimming, the fish were fed in 8 dpf in a 6-well plate. The larvae were sedated 1 h after feeding. After removing the leftover paramecia by washing twice, groups of five larvae were placed in an anesthetic solution in a 96-well round-bottom black plate. An enzyme-linked immunosorbent assay reader (Bio-Tek Synergy HT in fluorescence area scan mode 9 × 9 multipoint/well, 0.1 s/point) with a fluorescein filter set was used to measure the intra-abdominal fluorescence signal (excitation wavelength, 485 nm; emission wavelength, 590 nm) [31].

2.8. Whole-Mount Oil Red O Staining

Zebrafish larvae were fixed overnight at 4 °C in PBS (GeneMark, Taichung, Taiwan) containing 4% paraformaldehyde. Wild-type (WT) and AgRP4.7NTR larvae were distributed in equal numbers into 2 mL tubes and were washed twice (10 min each) with PBS. The larvae were pre-stained with 60% isopropyl alcohol for at least one hour. Thereafter, the larvae were stained with 0.5% Oil Red O solution (Sigma-Aldrich, Burlington, MA, USA) for two hours at 4 °C and then rinsed in PBS. The larvae were observed using a bright-field dissecting microscope (Stemi 305; Carl Zeiss AG, Oberkochen, Germany) while being preserved in 70% glycerol.

2.9. Statistical Analysis

Except for the quantitative body length data in the blind test experiment, which were shown as box plots, all the other data are presented as the mean ± standard error of the mean (SEM). For statistical purposes, T-TEST analysis was used. All analyses were conducted using the GraphPad Prism 9.0 program (GraphPad, San Diego, CA, USA). Differences were considered statistically significant at p < 0.05.

3. Results

3.1. Generation of Transgenic AgRP4.7NTR Zebrafish Lines

Firstly, we amplified 4 distinct lengths of the 5’ flanking region of the agrp gene from the genomic PCR amplicons: AgRP (2.7K), AgRP (3.4K), AgRP (4.0K), and AgRP (4.7K). Then, we created four agrp-EGFP-NTR constructs and used them to generate germline-transmissible transgenic zebrafish lines to verify the tissue specificity of the agrp promoter and achieve permanent green fluorescence expression in zebrafish AgRP neurons (Figure 1A).
The transgenic fish lines were established following the general Tol2 transposon protocols [42]. The implanted embryos were microscopically evaluated at 36 hpf, assessed for their fluorescence rate (Table 1), photographed (Figure 1B), reared to sexual maturity, and screened for prospective founders. The fluorescence of the five dpf progeny of the founder fish was examined and evaluated using microscopy after they were mated with WT fish.
The transient expression ratio of EGFP in 36 hpf larvae was determined by injecting embryos with constructs containing the 5’ flanking region of different lengths of the AgRP promoter. Embryos injected with the AgRP (4.7K), AgRP (4.0K), and AgRP (3.4K) constructs showed EGFP expression in the anterior region of the brain or trunk of the larvae (Figure 1B). The percentage of fluorescence ratio in AgRP (4.7K) was considerably higher than that of the other groups (Table 1). None of the embryos injected with AgRP (2.7K) showed EGFP expression. This suggested an insufficient length of the promoter region for transcriptional factors to bind in this group. Moreover, the AgRP (4.7K) promoter-driven transgenic construct (AgRP4.7NTR) has become the most promising form that could produce functional transgenic fish with stable EGFP-NTR fluorescence in the AgRP neurons of zebrafish.

3.2. EGFP-NTR Fusion Protein Causes Target Neural Cell Ablation

We adopted a previously reported NTR cell ablation system in this study [26,43]. We fused EGFP with bacterial NTR to form the EGFP-NTR gene fusion, speculating that its expression would not only produce green fluorescence but also have NTR activity to induce cytotoxicity via MTZ treatment. To confirm EGFP-NTR protein activity in vivo, we used the ubiquitous β-actin promoter to drive the EGFP-NTR fusion protein and performed a microinjection test in fertilized zebrafish embryos (Figure S1A). The larvae with this construct showed a green signal in muscle tissues; however, the green fluorescence was quenched by MTZ treatment (Figure S1B).
To ensure the capability of the NTR cell ablation system for neuronal cell ablation, the POMCNTR and AgRP4.7NTR transgenic fish experiments were performed (Figure 2A). After three days of MTZ treatment beginning at 2 dpf, the green fluorescence signals of POMC and AgRP neurons in 5 dpf larvae were quenched (Figure 2B,D). WISH analyses revealed significantly decreased expression patterns of pomc and agrp compared to those in WT fish (Figure 2C,E). As a result, the POMC and AgRP neurons were efficiently ablated by the NTR cell ablation system. Moreover, the AgRP4.7NTR transgenic fish line had a distinct fluorescence pattern that matched the site of AgRP neuron fluorescence [36] or the in situ hybridization pattern [44], as described in the literature.

3.3. The 4.7 kb Zebrafish agrp Promoter Has Several Essential Transcription Factor-Binding Sites That Are Exclusively Expressed in the AgRP Neuron

Since no information was available on the transcription factor datasheet for the zebrafish agrp promoter in the literature, we used the Table Browser website “https://genome.ucsc.edu/cgi-bin/hgTables” (accessed on 20 October 2022) to search the 5′ flanking region of the transcription start site (4749 bp in length) of the agrp gene in the zebrafish chromosome position: chr7: 35,083,136–35,087,884. As a result, many potential transcription factor-binding sites were identified in the 4.7 kb agrp upstream region. In the proximal region (Figure 3A), several predicted sites for developmental regulatory factors were found, including consensus motifs of brain-specific homeobox (Bsx) [18,45,46,47], GATA-binding protein 1 (Gata1) [48], forkhead box O1 (Foxo1) [18,49], nuclear receptor subfamily 3 group C member 1 (Nr3c1) [45,46], Kruppel-like factor 4 (Klf4) [50,51,52], and ISL LIM homeobox 1 (Isl1) [45,53], all of which are considered key AgRP transcription factors or enhancers in mice and humans.
As a result, we surmised that the 4.0–4.7 kb promoter region is an important regulatory area of the agrp promoter (Figure 3B), regulating the essential promoter activity of agrp, including the consensus motifs of cAMP-responsive element-binding protein 1 [18,51], Gata1, Foxo1, and Isl1. The findings of this study suggest that this region may play an essential role in enhancing agrp gene expression (Figure 3C).

3.4. Targeted Ablation of AgRP Neuron Represses Food Intake and Body Growth in Zebrafish Larvae

A blind test experiment was performed to validate whether the POMCNTR and AgRP4.7NTR transgenic fish can deplete POMC or AgRP neurons under MTZ treatment to accomplish neuronal ablation and further impact appetite control. In the experiment, 2 fish groups were set up, each with 25 POMCNTR, AgRP4.7NTR, and WT fish for mixed feeding. During the experiment, one control group received no medication. In contrast, the other experimental group received 1 mM MTZ beginning at 1 week of age, and the drug was constantly administered for 4 weeks until the fifth week, followed by a halt for 1 week until the sixth week (Figure 4A).
The two groups of experimental fish were first observed using a fluorescence microscope to differentiate POMCNTR, AgRP4.7NTR, and WT. For each group, the survival rate (Figure 4B) and body length were measured. The values of both parameters were calculated and plotted as a statistical graph (Figure 4C). In the experimental group treated with MTZ, POMCNTR fish had a larger body size, while AgRP4.7NTR fish were relatively thin and small.
Next, we employed a previously established nursing regimen to generate fluorescent food, consisting of larval fish meal with lipophilic tracer 4-Di-10-ASP-labeled paramecia [31], to evaluate the pattern of food consumption at the larval stage of the MTZ-treated AgRP4.7NTR. The larvae consumed luminous food, which caused strong fluorescence signals to build up in their gut (Figure 5A).
At 8 dpf, the AgRP4.7NTR larvae treated with MTZ showed anorexia, with a 0.44-fold decrease in food consumption compared with WT zebrafish (Figure 5B). Next, we investigated whether growth and energy budget were related to food intake in AgRP4.7NTR and discovered that contemporaneous observations were required to estimate these parameters. Moreover, AgRP4.7NTR transgenic fish showed a considerable decrease in body length (Figure 6A,B). Furthermore, the accumulation of lipid droplets in MTZ-treated AgRP4.7NTR was substantially lower than that in WT larvae (Figure 6C,D).

3.5. Defective AgRP Neuron Suppresses the Expression of Orexigenic Genes and Growth Hormones While Increasing the Anorexigenic Gene Expression in Zebrafish Larvae

Appetite-related gene expression levels in the brains after AgRP neuron ablation were determined using real-time RT-qPCR to analyze the brain gene profiles of MTZ-treated AgRP4.7NTR larvae. To compare the gene expression ratio of appetite-related genes between MTZ-treated WT and AgRP4.7NTR larvae, we examined the expression levels of several critical factors, including orexigenic genes, agrp, agrp2, npy [54,55], prodynorphin (pdyn) [56,57], hypothalamic hypocretin/orexin (hcrt) [58], and galanin/GMAP prepropeptide (gal) [59], which were repressed by AgRP neuron ablation (Figure 7A). In contrast, the levels of anorexigenic genes, including brain-derived neurotrophic factor (bdnf) [60,61], single-minded homolog 1-a (sim1a) [57,62], thyrotropin-releasing hormone (trh) [57,63], thyroglobulin (tg) [63], and arginine vasopressin (avp) [64,65], were promoted by AgRP neuron ablation (Figure 7B). Additionally, the expression levels of growth hormone (GH)/insulin-like growth factor (IGF) axis genes, including gh1 [23,29,31], growth hormone receptor a (ghra), ghrb, igf1, igf2b, insulin-like growth factor-binding protein 1b (igfbp1b), igfbp5b, igfbp6a, and igfbp6b, were decreased by AgRP neuron ablation (Figure 7C).
WISH was used to identify the appetite-related gene expression patterns and visible expression of genes, including npy, bdnf, trh, tg, and gh1, and the results were consistent with those of real-time RT-qPCR (Figure 7D). Our data showed that most orexigenic and growth hormone mRNA levels were significantly lower in MTZ-treated AgRP4.7NTR than in WT at 8 dpf, and most anorexigenic mRNA levels were markedly higher in MTZ-treated AgRP4.7NTR than in WT at 8 dpf. These results showed that our newly established MTZ-treated AgRP4.7NTR larvae could effectively ablate AgRP neurons, reduce the AgRP neuron peptide signal, and further affect the regulation of downstream brain appetite-related genes, resulting in appetite and growth suppression (Figure 8).

4. Discussion

To the best of our knowledge, no stable transgenic lines harboring agrp gene promoters have been described previously, and none of the AgRP-specific promoters in zebrafish have been subjected to an in-depth analysis so far. The development of transgenic zebrafish expressing a reporter gene in specific tissues or organs will aid in the study of vertebrate organogenesis, as zebrafish have gained popularity as model organisms. agrp gene expression in AgRP neurons in zebrafish has previously been demonstrated [44]. The zebrafish agrp promoter region was identified and described in this study.
In this study, we successfully cloned ARC-specific promoters containing either pomc [39] or agrp and generated transgenic zebrafish with ARC-specific promoter-driven transgenes. Following MTZ treatment of transgenic larvae, the target cells began to undergo apoptosis. For different promoters, such as β-actin, pomc, and agrp, fluorescence signal intensity in the target cell decreased and eventually disappeared. This finding demonstrated that the EGFP-NTR fusion protein can indicate the target area with visible fluorescence and retain cell toxicity. Therefore, it can potentially be used in a cell ablation technique in the future.
Transcription factor-binding site analysis of the 5’ flanking region of the transcription start site of the agrp gene showed that several predicted transcription factors and enhancers were involved in the promoter region (–591 to –1 bp and –4749 to –4149 bp). Previous literature suggests that among the transcription factors we identified, ISL1, BSX, and glucocorticoid receptor (GR, known as Nr3c1) bind to the homeodomain response elements (HDREs) and glucocorticoid response element (GRE) to form a ternary complex, which then synergistically stimulates glucocorticoid-directed transactivation of the agrp promoter, likely by facilitating the recruitment of transcriptional coactivators to the agrp promoter [45]. This hypothesis is also consistent with the findings of the present study.
We also discovered that appetite inhibition in the MTZ-treated AgRP4.7NTR model was associated with decreased hunger signals and inhibited somatic growth, as indicated by the gene profile results in this study. Moreover, we have summarized the importance of melanocortin circuits in controlling hunger. Our results contribute to a better understanding of the primary processes relating AgRP signaling and energy intake in zebrafish.
As a result, the 4.7 kb agrp promoter identified in this research will be a powerful tool for in vivo studies. It will make it easy for researchers to set up zebrafish models of human metabolic syndromes, such as appetite disorders and obesity, to find novel target genes involved in appetite regulation and to test medications for treating these disorders.
In addition to mimicking human Prader–Willi syndrome or bdelygmia by affecting the regulatory centers of feeding behavior, we can assess metabolism-related genes, such as peroxisome proliferator-activated receptor gamma [66], diacylglycerol O-acyltransferase 2 [67], and CCAAT enhancer-binding protein alpha [68], and simulate and investigate obesity induced by various factors by overexpressing or knocking out genes in AgRP neurons to identify effective strategies to maintain health, through methods such as drug screening and gene therapy.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines11020499/s1, Figure S1: Functional test of EGFP-NTR fusion protein, Table S1: Primer sequences used for real-time quantitative polymerase chain reaction (qPCR), Table S2: Primer sequences used for in situ hybridization probes.

Author Contributions

Methodology, G.M.H., K.-Y.Y. and C.-Y.L.; Writing—Original Draft Preparation, C.-Y.L.; Investigation, C.-Y.L. and H.-H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Science and Technology Council, Taiwan (grant number NSTC 111-2313-B-A49-002- to Guor Mour Her) and the Chang-Chung Memorial Hospital (grant number CORPG2M0071 to Kun-Yun Yeh).

Institutional Review Board Statement

Animal experiments in this study were carried out with the approval and as per the guidelines of the Institutional Animal Care and Use Committee (approval ID: 1110209) of the National Yang Ming Chiao Tung University, Taipei 112, Taiwan.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Funahashi, H.; Hori, T.; Shimoda, Y.; Mizushima, H.; Ryushi, T.; Katoh, S.; Shioda, S. Morphological evidence for neural interactions between leptin and orexin in the hypothalamus. Regul. Pept. 2000, 92, 31–35. [Google Scholar] [CrossRef] [PubMed]
  2. Wilding, J.P. Neuropeptides and appetite control. Diabet. Med. 2002, 19, 619–627. [Google Scholar] [CrossRef] [PubMed]
  3. Coll, A.P.; Farooqi, I.S.; O’Rahilly, S. The hormonal control of food intake. Cell 2007, 129, 251–262. [Google Scholar] [CrossRef] [PubMed]
  4. Wu, Q.; Palmiter, R.D. GABAergic signaling by AgRP neurons prevents anorexia via a melanocortin-independent mechanism. Eur. J. Pharmacol. 2011, 660, 21–27. [Google Scholar] [CrossRef] [PubMed]
  5. Friedman, J.M. Leptin and the endocrine control of energy balance. Nat. Metab. 2019, 1, 754–764. [Google Scholar] [CrossRef]
  6. Cheung, C.C.; Clifton, D.K.; Steiner, R.A. Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology 1997, 138, 4489–4492. [Google Scholar] [CrossRef]
  7. Cowley, M.A.; Smart, J.L.; Rubinstein, M.; Cerdan, M.G.; Diano, S.; Horvath, T.L.; Cone, R.D.; Low, M.J. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001, 411, 480–484. [Google Scholar] [CrossRef]
  8. Konner, A.C.; Janoschek, R.; Plum, L.; Jordan, S.D.; Rother, E.; Ma, X.; Xu, C.; Enriori, P.; Hampel, B.; Barsh, G.S.; et al. Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production. Cell Metab. 2007, 5, 438–449. [Google Scholar] [CrossRef]
  9. Broberger, C.; Johansen, J.; Johansson, C.; Schalling, M.; Hokfelt, T. The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and monosodium glutamate-treated mice. Proc. Natl. Acad. Sci. USA 1998, 95, 15043–15048. [Google Scholar] [CrossRef]
  10. Shutter, J.R.; Graham, M.; Kinsey, A.C.; Scully, S.; Luthy, R.; Stark, K.L. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in obese and diabetic mutant mice. Genes Dev. 1997, 11, 593–602. [Google Scholar] [CrossRef] [Green Version]
  11. Ollmann, M.M.; Wilson, B.D.; Yang, Y.K.; Kerns, J.A.; Chen, Y.; Gantz, I.; Barsh, G.S. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 1997, 278, 135–138. [Google Scholar] [CrossRef] [PubMed]
  12. Werther, G.A.; Hogg, A.; Oldfield, B.J.; McKinley, M.J.; Figdor, R.; Allen, A.M.; Mendelsohn, F.A. Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology 1987, 121, 1562–1570. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef] [PubMed]
  14. Sternson, S.M.; Atasoy, D. Agouti-related protein neuron circuits that regulate appetite. Neuroendocrinology 2014, 100, 95–102. [Google Scholar] [CrossRef] [PubMed]
  15. Vong, L.; Ye, C.; Yang, Z.; Choi, B.; Chua, S., Jr.; Lowell, B.B. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 2011, 71, 142–154. [Google Scholar] [CrossRef] [PubMed]
  16. Enriori, P.J.; Evans, A.E.; Sinnayah, P.; Jobst, E.E.; Tonelli-Lemos, L.; Billes, S.K.; Glavas, M.M.; Grayson, B.E.; Perello, M.; Nillni, E.A.; et al. Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons. Cell Metab. 2007, 5, 181–194. [Google Scholar] [CrossRef] [PubMed]
  17. Creemers, J.W.; Pritchard, L.E.; Gyte, A.; Le Rouzic, P.; Meulemans, S.; Wardlaw, S.L.; Zhu, X.; Steiner, D.F.; Davies, N.; Armstrong, D.; et al. Agouti-related protein is posttranslationally cleaved by proprotein convertase 1 to generate agouti-related protein (AGRP)83-132: Interaction between AGRP83-132 and melanocortin receptors cannot be influenced by syndecan-3. Endocrinology 2006, 147, 1621–1631. [Google Scholar] [CrossRef]
  18. Zagmutt, S.; Mera, P.; Soler-Vazquez, M.C.; Herrero, L.; Serra, D. Targeting AgRP neurons to maintain energy balance: Lessons from animal models. Biochem. Pharmacol. 2018, 155, 224–232. [Google Scholar] [CrossRef]
  19. Ollmann, M.M.; Lamoreux, M.L.; Wilson, B.D.; Barsh, G.S. Interaction of Agouti protein with the melanocortin 1 receptor in vitro and in vivo. Genes Dev. 1998, 12, 316–330. [Google Scholar] [CrossRef]
  20. Yeo, G.S.H.; Chao, D.H.M.; Siegert, A.M.; Koerperich, Z.M.; Ericson, M.D.; Simonds, S.E.; Larson, C.M.; Luquet, S.; Clarke, I.; Sharma, S.; et al. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol. Metab. 2021, 48, 101206. [Google Scholar] [CrossRef]
  21. Yeo, G.S.; Heisler, L.K. Unraveling the brain regulation of appetite: Lessons from genetics. Nat. Neurosci. 2012, 15, 1343–1349. [Google Scholar] [CrossRef] [PubMed]
  22. Baldini, G.; Phelan, K.D. The melanocortin pathway and control of appetite-progress and therapeutic implications. J. Endocrinol. 2019, 241, R1–R33. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, C.; Forlano, P.M.; Cone, R.D. AgRP and POMC neurons are hypophysiotropic and coordinately regulate multiple endocrine axes in a larval teleost. Cell Metab. 2012, 15, 256–264. [Google Scholar] [CrossRef]
  24. Qian, S.; Chen, H.; Weingarth, D.; Trumbauer, M.E.; Novi, D.E.; Guan, X.; Yu, H.; Shen, Z.; Feng, Y.; Frazier, E.; et al. Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Mol. Cell. Biol. 2002, 22, 5027–5035. [Google Scholar] [CrossRef] [PubMed]
  25. Wortley, K.E.; Anderson, K.D.; Yasenchak, J.; Murphy, A.; Valenzuela, D.; Diano, S.; Yancopoulos, G.D.; Wiegand, S.J.; Sleeman, M.W. Agouti-related protein-deficient mice display an age-related lean phenotype. Cell Metab. 2005, 2, 421–427. [Google Scholar] [CrossRef]
  26. Wu, Q.; Howell, M.P.; Palmiter, R.D. Ablation of neurons expressing agouti-related protein activates fos and gliosis in postsynaptic target regions. J. Neurosci. 2008, 28, 9218–9226. [Google Scholar] [CrossRef]
  27. Wu, Q.; Whiddon, B.B.; Palmiter, R.D. Ablation of neurons expressing agouti-related protein, but not melanin concentrating hormone, in leptin-deficient mice restores metabolic functions and fertility. Proc. Natl. Acad. Sci. USA 2012, 109, 3155–3160. [Google Scholar] [CrossRef]
  28. Shainer, I.; Michel, M.; Marquart, G.D.; Bhandiwad, A.A.; Zmora, N.; Ben-Moshe Livne, Z.; Zohar, Y.; Hazak, A.; Mazon, Y.; Forster, D.; et al. Agouti-Related Protein 2 Is a New Player in the Teleost Stress Response System. Curr. Biol. 2019, 29, 2009–2019.e7. [Google Scholar] [CrossRef]
  29. Lohr, H.; Hess, S.; Pereira, M.M.A.; Reinoss, P.; Leibold, S.; Schenkel, C.; Wunderlich, C.M.; Kloppenburg, P.; Bruning, J.C.; Hammerschmidt, M. Diet-Induced Growth Is Regulated via Acquired Leptin Resistance and Engages a Pomc-Somatostatin-Growth Hormone Circuit. Cell Rep. 2018, 23, 1728–1741. [Google Scholar] [CrossRef]
  30. Song, Y.; Cone, R.D. Creation of a genetic model of obesity in a teleost. FASEB J. 2007, 21, 2042–2049. [Google Scholar] [CrossRef] [Green Version]
  31. Hsieh, Y.W.; Tsai, Y.W.; Lai, H.H.; Lai, C.Y.; Lin, C.Y.; Her, G.M. Depletion of Alpha-Melanocyte-Stimulating Hormone Induces Insatiable Appetite and Gains in Energy Reserves and Body Weight in Zebrafish. Biomedicines 2021, 9, 941. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, Q.; Liu, C.; Uchida, A.; Chuang, J.C.; Walker, A.; Liu, T.; Osborne-Lawrence, S.; Mason, B.L.; Mosher, C.; Berglund, E.D.; et al. Arcuate AgRP neurons mediate orexigenic and glucoregulatory actions of ghrelin. Mol. Metab. 2014, 3, 64–72. [Google Scholar] [CrossRef] [PubMed]
  33. Luquet, S.; Perez, F.A.; Hnasko, T.S.; Palmiter, R.D. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 2005, 310, 683–685. [Google Scholar] [CrossRef] [PubMed]
  34. Suster, M.L.; Abe, G.; Schouw, A.; Kawakami, K. Transposon-mediated BAC transgenesis in zebrafish. Nat. Protoc. 2011, 6, 1998–2021. [Google Scholar] [CrossRef] [PubMed]
  35. Suster, M.L.; Sumiyama, K.; Kawakami, K. Transposon-mediated BAC transgenesis in zebrafish and mice. BMC Genom. 2009, 10, 477. [Google Scholar] [CrossRef]
  36. Shainer, I.; Buchshtab, A.; Hawkins, T.A.; Wilson, S.W.; Cone, R.D.; Gothilf, Y. Novel hypophysiotropic AgRP2 neurons and pineal cells revealed by BAC transgenesis in zebrafish. Sci. Rep. 2017, 7, 44777. [Google Scholar] [CrossRef]
  37. Hagerman, G.F.; Noel, N.C.; Cao, S.Y.; DuVal, M.G.; Oel, A.P.; Allison, W.T. Rapid Recovery of Visual Function Associated with Blue Cone Ablation in Zebrafish. PLoS ONE 2016, 11, e0166932. [Google Scholar] [CrossRef]
  38. Stoddard, M.; Huang, C.; Enyedi, B.; Niethammer, P. Live imaging of leukocyte recruitment in a zebrafish model of chemical liver injury. Sci. Rep. 2019, 9, 28. [Google Scholar] [CrossRef]
  39. Liu, N.A.; Huang, H.; Yang, Z.; Herzog, W.; Hammerschmidt, M.; Lin, S.; Melmed, S. Pituitary corticotroph ontogeny and regulation in transgenic zebrafish. Mol. Endocrinol. 2003, 17, 959–966. [Google Scholar] [CrossRef]
  40. Jowett, T. Double in situ hybridization techniques in zebrafish. Methods 2001, 23, 345–358. [Google Scholar] [CrossRef]
  41. Shimada, Y.; Hirano, M.; Nishimura, Y.; Tanaka, T. A high-throughput fluorescence-based assay system for appetite-regulating gene and drug screening. PLoS ONE 2012, 7, e52549. [Google Scholar] [CrossRef] [PubMed]
  42. Suster, M.L.; Kikuta, H.; Urasaki, A.; Asakawa, K.; Kawakami, K. Transgenesis in zebrafish with the tol2 transposon system. Methods Mol. Biol. 2009, 561, 41–63. [Google Scholar] [CrossRef] [PubMed]
  43. Curado, S.; Stainier, D.Y.; Anderson, R.M. Nitroreductase-mediated cell/tissue ablation in zebrafish: A spatially and temporally controlled ablation method with applications in developmental and regeneration studies. Nat. Protoc. 2008, 3, 948–954. [Google Scholar] [CrossRef]
  44. Song, Y.; Golling, G.; Thacker, T.L.; Cone, R.D. Agouti-related protein (AGRP) is conserved and regulated by metabolic state in the zebrafish, Danio rerio. Endocrine 2003, 22, 257–265. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, B.; Lee, S.; Lee, S.K.; Lee, J.W. The LIM-homeobox transcription factor Isl1 plays crucial roles in the development of multiple arcuate nucleus neurons. Development 2016, 143, 3763–3773. [Google Scholar] [CrossRef]
  46. Lee, B.; Kim, S.G.; Kim, J.; Choi, K.Y.; Lee, S.; Lee, S.K.; Lee, J.W. Brain-specific homeobox factor as a target selector for glucocorticoid receptor in energy balance. Mol. Cell. Biol. 2013, 33, 2650–2658. [Google Scholar] [CrossRef]
  47. Schredelseker, T.; Veit, F.; Dorsky, R.I.; Driever, W. Bsx Is Essential for Differentiation of Multiple Neuromodulatory Cell Populations in the Secondary Prosencephalon. Front. Neurosci. 2020, 14, 525. [Google Scholar] [CrossRef]
  48. Deng, W.; Lee, J.; Wang, H.; Miller, J.; Reik, A.; Gregory, P.D.; Dean, A.; Blobel, G.A. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 2012, 149, 1233–1244. [Google Scholar] [CrossRef]
  49. Kitamura, T.; Feng, Y.; Kitamura, Y.I.; Chua, S.C., Jr.; Xu, A.W.; Barsh, G.S.; Rossetti, L.; Accili, D. Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat. Med. 2006, 12, 534–540. [Google Scholar] [CrossRef]
  50. Imbernon, M.; Sanchez-Rebordelo, E.; Gallego, R.; Gandara, M.; Lear, P.; Lopez, M.; Dieguez, C.; Nogueiras, R. Hypothalamic KLF4 mediates leptin’s effects on food intake via AgRP. Mol. Metab. 2014, 3, 441–451. [Google Scholar] [CrossRef]
  51. Nakajima, K.; Cui, Z.; Li, C.; Meister, J.; Cui, Y.; Fu, O.; Smith, A.S.; Jain, S.; Lowell, B.B.; Krashes, M.J.; et al. Gs-coupled GPCR signalling in AgRP neurons triggers sustained increase in food intake. Nat. Commun. 2016, 7, 10268. [Google Scholar] [CrossRef] [PubMed]
  52. Ilnytska, O.; Stutz, A.M.; Park-York, M.; York, D.A.; Ribnicky, D.M.; Zuberi, A.; Cefalu, W.T.; Argyropoulos, G. Molecular mechanisms for activation of the agouti-related protein and stimulation of appetite. Diabetes 2011, 60, 97–106. [Google Scholar] [CrossRef] [PubMed]
  53. Nasif, S.; de Souza, F.S.; Gonzalez, L.E.; Yamashita, M.; Orquera, D.P.; Low, M.J.; Rubinstein, M. Islet 1 specifies the identity of hypothalamic melanocortin neurons and is critical for normal food intake and adiposity in adulthood. Proc. Natl. Acad. Sci. USA 2015, 112, E1861–E1870. [Google Scholar] [CrossRef] [PubMed]
  54. Yokobori, E.; Azuma, M.; Nishiguchi, R.; Kang, K.S.; Kamijo, M.; Uchiyama, M.; Matsuda, K. Neuropeptide Y stimulates food intake in the Zebrafish, Danio rerio. J. Neuroendocrinol. 2012, 24, 766–773. [Google Scholar] [CrossRef] [PubMed]
  55. Mercer, R.E.; Chee, M.J.; Colmers, W.F. The role of NPY in hypothalamic mediated food intake. Front. Neuroendocrinol. 2011, 32, 398–415. [Google Scholar] [CrossRef] [PubMed]
  56. Garfield, A.S.; Shah, B.P.; Burgess, C.R.; Li, M.M.; Li, C.; Steger, J.S.; Madara, J.C.; Campbell, J.N.; Kroeger, D.; Scammell, T.E.; et al. Dynamic GABAergic afferent modulation of AgRP neurons. Nat. Neurosci. 2016, 19, 1628–1635. [Google Scholar] [CrossRef]
  57. Krashes, M.J.; Shah, B.P.; Madara, J.C.; Olson, D.P.; Strochlic, D.E.; Garfield, A.S.; Vong, L.; Pei, H.; Watabe-Uchida, M.; Uchida, N.; et al. An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature 2014, 507, 238–242. [Google Scholar] [CrossRef]
  58. Merkle, F.T.; Maroof, A.; Wataya, T.; Sasai, Y.; Studer, L.; Eggan, K.; Schier, A.F. Generation of neuropeptidergic hypothalamic neurons from human pluripotent stem cells. Development 2015, 142, 633–643. [Google Scholar] [CrossRef]
  59. Qualls-Creekmore, E.; Yu, S.; Francois, M.; Hoang, J.; Huesing, C.; Bruce-Keller, A.; Burk, D.; Berthoud, H.R.; Morrison, C.D.; Munzberg, H. Galanin-Expressing GABA Neurons in the Lateral Hypothalamus Modulate Food Reward and Noncompulsive Locomotion. J. Neurosci. 2017, 37, 6053–6065. [Google Scholar] [CrossRef]
  60. Benarroch, E.E. Brain-derived neurotrophic factor: Regulation, effects, and potential clinical relevance. Neurology 2015, 84, 1693–1704. [Google Scholar] [CrossRef]
  61. Marosi, K.; Mattson, M.P. BDNF mediates adaptive brain and body responses to energetic challenges. Trends Endocrinol. Metab. 2014, 25, 89–98. [Google Scholar] [CrossRef] [PubMed]
  62. Xi, D.; Roizen, J.; Lai, M.; Gandhi, N.; Kublaoui, B. Paraventricular nucleus Sim1 neuron ablation mediated obesity is resistant to high fat diet. PLoS ONE 2013, 8, e81087. [Google Scholar] [CrossRef] [PubMed]
  63. Mullur, R.; Liu, Y.Y.; Brent, G.A. Thyroid hormone regulation of metabolism. Physiol. Rev. 2014, 94, 355–382. [Google Scholar] [CrossRef]
  64. Pei, H.; Sutton, A.K.; Burnett, K.H.; Fuller, P.M.; Olson, D.P. AVP neurons in the paraventricular nucleus of the hypothalamus regulate feeding. Mol. Metab. 2014, 3, 209–215. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, A.; Madara, J.C.; Wu, C.; Andermann, M.L.; Lowell, B.B. Neural basis for regulation of vasopressin secretion by anticipated disturbances in osmolality. eLife 2021, 10, e66609. [Google Scholar] [CrossRef]
  66. Harrington, W.W.; Britt, C.S.; Wilson, J.G.; Milliken, N.O.; Binz, J.G.; Lobe, D.C.; Oliver, W.R.; Lewis, M.C.; Ignar, D.M. The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice. PPAR Res. 2007, 2007, 97125. [Google Scholar] [CrossRef]
  67. Chen, H.C.; Farese, R.V., Jr. Inhibition of triglyceride synthesis as a treatment strategy for obesity: Lessons from DGAT1-deficient mice. Arterioscler Thromb. Vasc. Biol. 2005, 25, 482–486. [Google Scholar] [CrossRef]
  68. Ruan, H.; Pownall, H.J.; Lodish, H.F. Troglitazone antagonizes tumor necrosis factor-alpha-induced reprogramming of adipocyte gene expression by inhibiting the transcriptional regulatory functions of NF-kappaB. J. Biol. Chem. 2003, 278, 28181–28192. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Transient expression analysis of the agrp-enhanced, green, fluorescent protein-nitroreductase (agrp-EGFP-NTR) deletion constructs. (A) Schematic diagram of the DNA construct used to generate AgRPNTR (Tg[agrp-EGFP-NTR]) transgenic zebrafish. AgRP (4.7K)-EGFP-NTR contains a 4749 bp 5′ flanking region of the agrp gene, and EGFP-NTR fusion has an in-frame 24 bp region downstream of the agrp translational start site. (B) Each deletion construct was generated as described in Section 2.2 for functional analyses of the promoter. A total of 4.6 nL (15 ng/µL) of the deletion constructs for (1) AgRP (2.7K)-EGFP-NTR, (2) AgRP (3.4K)-EGFP-NTR, and (3) AgRP (4.0K)-EGFP-NTR and the full-length construct for (4) AgRP (4.7K)-EGFP-NTR were microinjected into one-cell stage embryos. EGFP signal was observed in 36 h post-fertilization (hpf) embryos injected with these constructs via fluorescence microscopy. Scale bar: 200 µm.
Figure 1. Transient expression analysis of the agrp-enhanced, green, fluorescent protein-nitroreductase (agrp-EGFP-NTR) deletion constructs. (A) Schematic diagram of the DNA construct used to generate AgRPNTR (Tg[agrp-EGFP-NTR]) transgenic zebrafish. AgRP (4.7K)-EGFP-NTR contains a 4749 bp 5′ flanking region of the agrp gene, and EGFP-NTR fusion has an in-frame 24 bp region downstream of the agrp translational start site. (B) Each deletion construct was generated as described in Section 2.2 for functional analyses of the promoter. A total of 4.6 nL (15 ng/µL) of the deletion constructs for (1) AgRP (2.7K)-EGFP-NTR, (2) AgRP (3.4K)-EGFP-NTR, and (3) AgRP (4.0K)-EGFP-NTR and the full-length construct for (4) AgRP (4.7K)-EGFP-NTR were microinjected into one-cell stage embryos. EGFP signal was observed in 36 h post-fertilization (hpf) embryos injected with these constructs via fluorescence microscopy. Scale bar: 200 µm.
Biomedicines 11 00499 g001
Figure 2. Establishment of POMCNTR and AgRP4.7NTR transgenic fish and neuron ablation tests. (A) Schematic diagram of the DNA construct used to generate POMCNTR and AgRP4.7NTR transgenic zebrafish lines. Fluorescence images of (1) POMCNTR and (3) AgRP4.7NTR larval head at 5 days post-fertilization (dpf) (Scale bar: 200 µm). Confocal images of (2) pro-opiomelanocortin (POMC) neurons of POMCNTR and (4) AgRP neurons of AgRP4.7NTR at 5 dpf (Scale bar: 20 µm). (B) Left: Fluorescence images of (1) POMCNTR transgenic zebrafish larvae at 5 dpf and (2) metronidazole (MTZ)-treated POMCNTR transgenic zebrafish larvae at 5 dpf, n = 3. Scale bar: 500 µm. Right: quantification diagram of fluorescence intensity. All values are the mean ± standard error of the mean (SEM), n = 30. *** p < 0.001. (C) Whole mount in situ hybridization (WISH) assays showing the expression signals of pomc transcripts in POMC neurons in wild-type (WT) and POMCNTR larvae at 5 dpf, n = 30. Scale bar: 100 µm. (D) Left: Fluorescence images of (1) AgRP4.7NTR transgenic zebrafish larvae at 5 dpf and (2) MTZ-treated AgRP4.7NTR transgenic zebrafish larvae at 5 dpf, n = 3. Scale bar: 500 µm. Right: quantification diagram of fluorescence intensity. All values reported as mean ± SEM, n = 30. ** p < 0.01. (E) WISH assay results showing the expression signals of agrp transcripts in AgRP neurons in WT and AgRP4.7NTR larvae at 5 dpf. Scale bar: 100 µm.
Figure 2. Establishment of POMCNTR and AgRP4.7NTR transgenic fish and neuron ablation tests. (A) Schematic diagram of the DNA construct used to generate POMCNTR and AgRP4.7NTR transgenic zebrafish lines. Fluorescence images of (1) POMCNTR and (3) AgRP4.7NTR larval head at 5 days post-fertilization (dpf) (Scale bar: 200 µm). Confocal images of (2) pro-opiomelanocortin (POMC) neurons of POMCNTR and (4) AgRP neurons of AgRP4.7NTR at 5 dpf (Scale bar: 20 µm). (B) Left: Fluorescence images of (1) POMCNTR transgenic zebrafish larvae at 5 dpf and (2) metronidazole (MTZ)-treated POMCNTR transgenic zebrafish larvae at 5 dpf, n = 3. Scale bar: 500 µm. Right: quantification diagram of fluorescence intensity. All values are the mean ± standard error of the mean (SEM), n = 30. *** p < 0.001. (C) Whole mount in situ hybridization (WISH) assays showing the expression signals of pomc transcripts in POMC neurons in wild-type (WT) and POMCNTR larvae at 5 dpf, n = 30. Scale bar: 100 µm. (D) Left: Fluorescence images of (1) AgRP4.7NTR transgenic zebrafish larvae at 5 dpf and (2) MTZ-treated AgRP4.7NTR transgenic zebrafish larvae at 5 dpf, n = 3. Scale bar: 500 µm. Right: quantification diagram of fluorescence intensity. All values reported as mean ± SEM, n = 30. ** p < 0.01. (E) WISH assay results showing the expression signals of agrp transcripts in AgRP neurons in WT and AgRP4.7NTR larvae at 5 dpf. Scale bar: 100 µm.
Biomedicines 11 00499 g002
Figure 3. The sequence and potential response elements in the 4.7 kb agrp promoter of zebrafish. The nucleotide sequence was downloaded from the Ensembl website “https://asia.ensembl.org/Danio_rerio/Info/Index” (accessed on 2 May 2022). (A) The translational start codon (ATG) is underlined and shown as position +1. Nucleotides are numbered on the right, starting with the translational codon. Potential binding motifs in the proximal region are also marked. (B) Sequences of the putative agrp regulatory element. The predicted binding motif within a distal region of agrp promoter. Potential binding motifs are marked. (C) Hypothetical model of agrp transcription activation in zebrafish. The proteins shown in the figure are: TATA-box-binding protein (Tbp), CCAAT/enhancer-binding protein alpha (Cebpα), ISL LIM homeobox 1 (Isl1), Kruppel-like factor 4 (Klf4), nuclear receptor subfamily 3 group C member 1 (Nr3c1), forkhead box O1 (Foxo1), GATA-binding protein 1 (Gata1), T cell acute lymphocytic leukemia 1 (Tal1), and brain-specific homeobox (Bsx).
Figure 3. The sequence and potential response elements in the 4.7 kb agrp promoter of zebrafish. The nucleotide sequence was downloaded from the Ensembl website “https://asia.ensembl.org/Danio_rerio/Info/Index” (accessed on 2 May 2022). (A) The translational start codon (ATG) is underlined and shown as position +1. Nucleotides are numbered on the right, starting with the translational codon. Potential binding motifs in the proximal region are also marked. (B) Sequences of the putative agrp regulatory element. The predicted binding motif within a distal region of agrp promoter. Potential binding motifs are marked. (C) Hypothetical model of agrp transcription activation in zebrafish. The proteins shown in the figure are: TATA-box-binding protein (Tbp), CCAAT/enhancer-binding protein alpha (Cebpα), ISL LIM homeobox 1 (Isl1), Kruppel-like factor 4 (Klf4), nuclear receptor subfamily 3 group C member 1 (Nr3c1), forkhead box O1 (Foxo1), GATA-binding protein 1 (Gata1), T cell acute lymphocytic leukemia 1 (Tal1), and brain-specific homeobox (Bsx).
Biomedicines 11 00499 g003
Figure 4. Blind test experiment of targeted neuron ablation transgenic fish. (A) Lateral views of POMCNTR, WT, and AgRP4.7NTR six-week-old larvae. (B) Survival rate of each group. (C) Statistical analysis of body length (jaw to caudal fin) in six-week-old POMCNTR, WT, and AgRP4.7NTR larvae. Data are shown as box plots including median (line), interquartile range (box), and 95% confidence interval (whiskers). *** p < 0.001.
Figure 4. Blind test experiment of targeted neuron ablation transgenic fish. (A) Lateral views of POMCNTR, WT, and AgRP4.7NTR six-week-old larvae. (B) Survival rate of each group. (C) Statistical analysis of body length (jaw to caudal fin) in six-week-old POMCNTR, WT, and AgRP4.7NTR larvae. Data are shown as box plots including median (line), interquartile range (box), and 95% confidence interval (whiskers). *** p < 0.001.
Biomedicines 11 00499 g004
Figure 5. Qualitative food intake assay for MTZ-treated WT and AgRP4.7NTR larvae. (A) Side views of 8 dpf larvae were observed under fluorescence lighting. MTZ-treated WT and AgRP4.7NTR larvae were cultivated with 4-Di-10-ASP-labeled paramecia for 1 h before observing the fluorescent contents in their guts. Scale bar: 2 mm. (B) Relationship between the relative amount of paramecia and fluorescence intensities of ingested paramecia in zebrafish. All values are represented as the mean ± SEM, n = 50. *** p < 0.001.
Figure 5. Qualitative food intake assay for MTZ-treated WT and AgRP4.7NTR larvae. (A) Side views of 8 dpf larvae were observed under fluorescence lighting. MTZ-treated WT and AgRP4.7NTR larvae were cultivated with 4-Di-10-ASP-labeled paramecia for 1 h before observing the fluorescent contents in their guts. Scale bar: 2 mm. (B) Relationship between the relative amount of paramecia and fluorescence intensities of ingested paramecia in zebrafish. All values are represented as the mean ± SEM, n = 50. *** p < 0.001.
Biomedicines 11 00499 g005
Figure 6. AgRP neuron ablation results in the inhibition of somatic growth and adipocyte formation. (A) Lateral view of MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf. Scale bar: 500 µm. (B) Statistics of body length measurement between MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf. All values are reported as mean ± SEM, n = 30. *** p < 0.001. (C) Lipid droplet accumulation was observed via whole-body Oil Red O staining of MTZ-treated WT and AgRP4.7NTR larvae at 21 dpf. Scale bar: 1 mm. (D) Left: Distribution of lipid droplets in MTZ-treated WT and AgRP4.7NTR larvae at 21 dpf, n = 30. Right: Intensity grading diagram.
Figure 6. AgRP neuron ablation results in the inhibition of somatic growth and adipocyte formation. (A) Lateral view of MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf. Scale bar: 500 µm. (B) Statistics of body length measurement between MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf. All values are reported as mean ± SEM, n = 30. *** p < 0.001. (C) Lipid droplet accumulation was observed via whole-body Oil Red O staining of MTZ-treated WT and AgRP4.7NTR larvae at 21 dpf. Scale bar: 1 mm. (D) Left: Distribution of lipid droplets in MTZ-treated WT and AgRP4.7NTR larvae at 21 dpf, n = 30. Right: Intensity grading diagram.
Biomedicines 11 00499 g006
Figure 7. Effects of AgRP ablation on appetite and growth regulation in the brain of zebrafish. Real-time RT-qPCR analysis was used to measure the mRNA expression levels of (A) orexigenic genes (agrp, agrp2, npy, pdyn, pro-melanin concentrating hormone (pmch), hcrt, gal, and neurotensin (nts)); (B) anorexigenic genes (bdnf, sim1a, trh, tg, avp, nucleobindin 1 (nucb1), corticotropin releasing hormone b (crhb), and oxytocin (oxt)); and (C) GH/IGF axis genes (gh1, ghra, ghrb, igf1, igf2b, igfbp1b, igfbp5b, igfbp6a, and igfbp6b) in MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf, n = 30. Values are represented as the mean ± S.E.M. *** p < 0.001. (D) WISH experiment revealed the expression levels and patterns of npy, bdnf, trh, tg, and gh1 in the brains of MTZ-treated WT and AgRP4.7NTR larvae at 5 dpf, n = 30. Scale bars = 100 µm.
Figure 7. Effects of AgRP ablation on appetite and growth regulation in the brain of zebrafish. Real-time RT-qPCR analysis was used to measure the mRNA expression levels of (A) orexigenic genes (agrp, agrp2, npy, pdyn, pro-melanin concentrating hormone (pmch), hcrt, gal, and neurotensin (nts)); (B) anorexigenic genes (bdnf, sim1a, trh, tg, avp, nucleobindin 1 (nucb1), corticotropin releasing hormone b (crhb), and oxytocin (oxt)); and (C) GH/IGF axis genes (gh1, ghra, ghrb, igf1, igf2b, igfbp1b, igfbp5b, igfbp6a, and igfbp6b) in MTZ-treated WT and AgRP4.7NTR larvae at 8 dpf, n = 30. Values are represented as the mean ± S.E.M. *** p < 0.001. (D) WISH experiment revealed the expression levels and patterns of npy, bdnf, trh, tg, and gh1 in the brains of MTZ-treated WT and AgRP4.7NTR larvae at 5 dpf, n = 30. Scale bars = 100 µm.
Biomedicines 11 00499 g007
Figure 8. Appetite-regulation pathway under AgRP neuron ablation. Schematic diagram of suppression of orexigenic factors (pdyn, hcrt, and gal) within the lateral hypothalamus area (LHA) and promotion of anorexigenic factors (bdnf, sim1a, trh, tg, and avp) within the paraventricular nucleus (PVN) via AgRP neuron ablation, leading to appetite and growth suppression.
Figure 8. Appetite-regulation pathway under AgRP neuron ablation. Schematic diagram of suppression of orexigenic factors (pdyn, hcrt, and gal) within the lateral hypothalamus area (LHA) and promotion of anorexigenic factors (bdnf, sim1a, trh, tg, and avp) within the paraventricular nucleus (PVN) via AgRP neuron ablation, leading to appetite and growth suppression.
Biomedicines 11 00499 g008
Table 1. Expression of EGFP in 36 hpf embryos injected with constructs containing the agrp promoter sequences.
Table 1. Expression of EGFP in 36 hpf embryos injected with constructs containing the agrp promoter sequences.
ConstructNo. of Embryo ObservedNo. of Embryo with EGFP ExpressionFluorescence Ratio (%)
HeadTrunkTotal
AgRP (2.7K)2050000.00
AgRP (3.4K)2151452.33
AgRP (4.0K)22156114.98
AgRP (4.7K)23232114318.53
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lin, C.-Y.; Yeh, K.-Y.; Lai, H.-H.; Her, G.M. AgRP Neuron-Specific Ablation Represses Appetite, Energy Intake, and Somatic Growth in Larval Zebrafish. Biomedicines 2023, 11, 499. https://doi.org/10.3390/biomedicines11020499

AMA Style

Lin C-Y, Yeh K-Y, Lai H-H, Her GM. AgRP Neuron-Specific Ablation Represses Appetite, Energy Intake, and Somatic Growth in Larval Zebrafish. Biomedicines. 2023; 11(2):499. https://doi.org/10.3390/biomedicines11020499

Chicago/Turabian Style

Lin, Chiu-Ya, Kun-Yun Yeh, Hsin-Hung Lai, and Guor Mour Her. 2023. "AgRP Neuron-Specific Ablation Represses Appetite, Energy Intake, and Somatic Growth in Larval Zebrafish" Biomedicines 11, no. 2: 499. https://doi.org/10.3390/biomedicines11020499

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop