Hypothalamic Menin regulates systemic rising old and cognitive decline
-
Loading metrics
Open Entry
Sight-reviewed
Overview Article
- Lige Leng,
- Ziqi Yuan,
- Xiao Su,
- Zhenlei Chen,
- Shangchen Yang,
- Meiqin Chen,
- Kai Zhuang,
- Hui Lin,
- Hao Solar,
- Huifang Li
x
- Published: March 16, 2023
- https://doi.org/10.1371/journal.pbio.3002033
Summary
Rising old is a systemic route of, which is a possibility component for impaired physiological capabilities, and at final demise. The molecular mechanisms riding rising old route of and the associated cognitive decline are no longer completely understood. The hypothalamus acts because the arbiter that orchestrates systemic rising old thru neuroinflammatory signaling. Our new findings revealed that Menin plays indispensable roles in neuroinflammation and brain vogue. Here, we discovered that the hypothalamic Menin signaling diminished in extinct mice, which correlates with systemic rising old and cognitive deficits. Restoring Menin expression in ventromedial nucleus of hypothalamus (VMH) of extinct mice extended lifespan, improved studying and memory, and ameliorated rising old biomarkers, while inhibiting Menin in VMH of center-extinct mice induced premature rising old and accelerated cognitive decline. We extra discovered that Menin epigenetically regulates neuroinflammatory and metabolic pathways, including D-serine metabolism. Rising old-associated Menin reduction ended in impaired D-serine release by VMH-hippocampus neural circuit, while D-serine complement rescued cognitive decline in extinct mice. Collectively, VMH Menin serves as a key regulator of systemic rising old and rising old-linked cognitive decline.
Citation: Leng L, Yuan Z, Su X, Chen Z, Yang S, Chen M, et al. (2023) Hypothalamic Menin regulates systemic rising old and cognitive decline. PLoS Biol 21(3):
e3002033.
https://doi.org/10.1371/journal.pbio.3002033
Academic Editor: Joseph Castellano, Icahn School of Remedy at Mount Sinai, UNITED STATES
Got: April 21, 2022; Permitted: February 9, 2023; Published: March 16, 2023
Copyright: © 2023 Leng et al. Here’s an launch secure admission to article disbursed below the terms of the Ingenious Commons Attribution License, which allows unrestricted spend, distribution, and reproduction in any medium, provided the long-established writer and supply are credited.
Knowledge Availability: All records generated or analyzed at some stage in this eye are integrated in this printed article (and its supplementary info info). The SRA accession quantity for RNA-seq records reported in this paper is: PRJNA783757 (SRR17042317- SRR17042322).
Funding: This work used to be supported by the Nationwide Natural Science Foundation of China (Grant: 81925010, 91849205, U1905207 and 92049202 to J.Z.; Grant: 81801337 and 82071520 to L.L.); The Nationwide Key Overview and Style Program of China (Grant: 2021YFA1101402 to J.Z.); The Traditional Overview Funds for the Central Universities (Grant: 20720190118 and 20720180049 to J.Z.); Xiamen Municipal Health Commission, Xiamen Municipal Bureau of Science and Abilities (3502Z20209005); Fujian Province Nature Science Foundation (Grant: 2019J05006 to L.L.); Xiamen Childhood Innovation Fund (Grant: 3502Z20206031 to L.L). The funders had no aim in eye develop, records sequence and diagnosis, determination to publish, or preparation of the manuscript.
Competing pursuits: The authors acquire declared that no competing pursuits exist.
Abbreviations:
ChIP,
chromatin immunoprecipitation; DAMP,
hurt-associated molecular pattern; DEG,
differentially expressed gene; GnRH,
gonadotropin-releasing hormone; HFS,
high-frequency stimulation; LPS,
lipopolysaccharide; LTP,
long-term potentiation; mEPSC,
little excitatory postsynaptic new; mIPSC,
little inhibitory postsynaptic new; MRM,
a couple of response monitoring; OPLS-DA,
orthogonal partial least-sq. discriminant diagnosis; PCA,
major component diagnosis; PHGDH,
phosphoglycerate dehydrogenase; PKA,
protein kinase A; PLS-DA,
partial least-sq. discriminant diagnosis; PRR,
pattern recognition receptor; PSPH,
phosphoserine phosphatase; PVDF,
polyvinylidene difluoride; SCT,
sucrose consumption test; SFA,
saturated fatty acid; SF-1,
steroidogenic component-1; SPF,
specific-pathogen-free; SPT,
sucrose preference test; SRM,
selected response monitoring; TLR4,
Toll-esteem receptor 4; TNF,
tumor necrosis component; VIP,
variable importance on projection; VMH,
ventromedial hypothalamus
Introduction
Rising old is characterized by the progressive and total deterioration of physiological capabilities, resulting within the stop of an organism’s lifespan [1–4]. Amongst brain and the sub brain situation, hypothalamus has been identified as excessive central regulators of rising old route of [5,6]. Particularly, neuroinflammatory IKKβ/NF-κB signaling in ventromedial hypothalamus (VMH) [5] is defined because the prance-regulator of systemic rising old. RelA (p65) phosphorylation, marker of NF-κB activation, will improve gradually at some stage in rising old. Activation of IKKβ/NF-κB signaling tunes down gonadotropin-releasing hormone (GnRH) release within the hypothalamus at some stage in rising old, while GnRH complement alleviates rising old-impaired neurogenesis and decelerates rising old.
Moreover rising old, VMH is excessive in regulating food intake and sustaining total-physique energy metabolic, glucose, and lipid balance [7]. VMH microinflammation has been tightly linked to the metabolic mode of macronutrients, especially the fatty acid-mediated oxidative metabolism [8,9]. Saturated fatty acids (SFAs) spark off Toll-esteem receptor 4 (TLR4) and its adaptor myeloid differentiation component 88 (MyD88) in VMH, resulting in activation of IKKβ/NF-κB [10,11], which attributable to this truth drove the expression of inflammatory genes. Then all yet again, the upstream molecules regulating neuroinflammation in VMH live unclear.
Hypothalamic tumor necrosis component (TNF) receptor mediates adaptive immunity primarily based on lipopolysaccharide (LPS), peptidoglycan and lipoteichoic acids, or hurt-associated molecular patterns (DAMPs) [12]. Immune cytokines, including interleukin-6 (IL-6) and IL-1β, are indispensable in inducing native inflammation [13]. Except for cytokine, TLRs represent a immense family of pattern recognition receptors (PRRs), amongst which TLR4 is discovered to be linked to obesity-linked hypothalamic inflammation [11]. Both NF-κB classical or weird and wonderful activation is mandatory for initiation, repairs, and development of hypothalamic inflammation [5,9]. Our new findings reported that a couple of endocrine neoplasia fashion 1 (MEN1; protein: Menin) pals with p65 to inhibit NF-κB transactivation [14], performing as a extremely indispensable component in inhibiting neuroinflammation. Therefore, we hypothesize that Menin may per chance per chance also act as upstream arbiter for rising old-associated neuroinflammation in VMH.
On this eye, we characterized the adjustments of Menin expression alongside with rising old in C57BL/6 mice. Then, we manipulated Menin levels selectively in steroidogenic component-1 (SF-1) neuron of VMH to ogle its capability effects on systemic rising old and cognitive capabilities. We then investigated the neuroinflammation cascades and cognitive adjustments underlying these circumstances. The outcomes explored a unique aim of Menin in regulating systemic rising old and cognitive aim.
Outcomes
Diminished Menin within the hypothalamus hastens systemic rising old
Inflammatory activation mediated by IKKβ/NF-κB within the hypothalamus used to be indispensable studied within the context of rising old, and extra induces the cognitive impairment associated with rising old [5]. In old eye, we discovered that Menin pals with p65 to inhibit nuclear component kappa-B (NF-κB) transactivation [14]. Therefore, we first examined the trends of Menin in 7 brain regions of young and feeble mice, and discovered that the decrease of Menin within the hypothalamus used to be essentially the most principal with age (Figs 1A–1D and S1A–S1L), which accompanies increased neuroinflammation within the hypothalamus (Fig 1E–1G). Next, by co-immunostaining Menin with SF-1, GFAP, and IBA1, we discovered that the expression of Menin diminished a good deal handiest in VMH SF-1 neurons nevertheless no longer in astrocytes and microglia in extinct mice brain (S1M–S1R Fig). These outcomes urged that the aim of Menin in SF-1 neuron may be strongly implicated in rising old. The expression of Menin gradually decreases with rising old, and is collected relatively high at 10M, when is take for subsequent experiments (S1S and S1T Fig). Earlier be taught acquire implicated SF-1 neurons, that are uncommon to hypothalamic VMH, as indispensable metabolic regulators [7]. VMH has been shown to protect watch over the repairs of energy homeostasis in total physique, VMH SF-1 neurons can answer to the nutritional situation [15–17]. To realise the prospective relevance of lowered Menin on hypothalamic neuroinflammation and systemic rising old, we generated steroidogenic component 1 (SF1) Cre Men1f/f conditional knockout mice (ScKO) by crossing SF1-Cre mice [18] with mice carrying floxed Men1 alleles (Men1f/f) [19] (Fig 1H). Menin deletion efficiency in SF-1 neurons used to be confirmed by western blotting, proper time-PCR, and immunostaining (Fig 1I–1L). Mice with SF-1 neuronal Menin knockdown show increased neuroinflammation in VMH (Fig 1M–1O). Menin deletion in SF-1 neuron attributable to this truth induced ScKO mice swear irregular metabolic circadian rhythm, increased food and water intake accompany with increased physique weight (Figs 1P–1T and S2B). ScKO mice are born at an expected mendelian frequency with a on the discipline of 1:1 intercourse ratio, without principal adjustments in physique weight, brain weight, and neuron quantity in hypothalamus and hippocampus (S2A–S2E Fig). Then all yet again, the lifespan in ScKO mice is a good deal diminished when when put next with manipulate animals in each and every male and female mice (Figs 1U and S3J). The ScKO mice furthermore swear rising old-linked phenotypes including lowered muscle fiber size, bone mass, pores and skin thickness, tail tendon collagen immoral-linking, clock genes expression, increased ventricular muscle thickness, and DNA methylation levels (Figs 1V–1Z and S2F–S2I). β-Gal staining on the liver, coronary heart, muscle, and hypothalamus of above mice indicated that the above tissues from ScKO mice had premature rising old (S2J–S2Q Fig). Moreover, cognitive decline because the indispensable rising old phenotype looked in 10-month-feeble male and female ScKO mice as measured with Morris water maze. ScKO mice showed impaired studying with increased secure away latency when put next with controls at some stage within the 6-day coaching section. Moreover, at some stage within the probe trial test, by which the hidden platform used to be eradicated on day 7, ScKO mice spent a good deal less time within the map quadrant. ScKO mice furthermore completed fewer entries into the platform procedure and wished a longer duration to commute from the entry designate the map zone when put next with controls. Moreover, ScKO mice furthermore showed lowered spontaneous alternations when put next with controls in T- and Y-maze assessments (Figs 1AA–1AE and S3N–S3U). We did now not ogle principal difference in locomotion, bother- and despair-linked habits assessments between protect watch over and ScKO mice (S3A–S3I, S3K–S3M, S3S, and S3T Figs). By electrophysiology, we discovered that VMH SF-1 neurons in ScKO mice showed diminished amplitude and frequency of sEPSC and sIPSC versus protect watch over mice (Fig 1AF and 1AG).
Fig 1. Diminished Menin within the hypothalamus hastens systemic rising old.
(A–C) Menin protein expression and mRNA levels within the hypothalamus of young (3M) and feeble (20M) mice; n = 4 mice. (D) Menin protein expression levels in 7 regions of young (3M) and feeble (20M) mice brain; n = 4 mice. Actin serves as a loading protect watch over. (E–G) Inflammatory components protein expression and mRNA levels within the hypothalamus of young (3M) and feeble (20M) mice; n = 3 mice. (H) Generation of conditional Men1-knockout mice by crossing Men1f/f mice with SF1-Cre mice (ScKO). (I–Okay) The protein and mRNA levels of Menin within the hypothalamus of 6 months ScKO and protect watch over mice; n = 3 mice. (L) Representative hypothalamus brain sections from 6 months protect watch over and ScKO mice stained with Menin (inexperienced) and SF1 (crimson) antibody. Piece used to be counterstained with DAPI (blue). Scale bar, 100 μm, 20 μm; n = 5 mice. (M–O) Inflammatory components protein expression and mRNA levels within the hypothalamus of ScKO (6M) and protect watch over (6M) mice; n = 3 mice. (P) Schematic diagram of launch circuit oblique calorimetry. (Q) Day and evening respiratory quotients (RER) of age-matched male ScKO mice (10M) and protect watch over mice (10M) were measured. Loyal dietary (R), water intake (S), and exercise (T) were furthermore measured. (U) Lifespan of those mice (Alter mice, n = 14; ScKO mice, n = 16). (V–Z) Male mice were sacrificed at 10 months for measuring muscle (quadriceps) fiber size (V), bone mass (2), dermal thickness (X, Y), and tail tendon breaking time (Z); n = 3~9 mice. Scale bar, 20 μm. (AA–AC) Ten months male ScKO and age-matched protect watch over mice habits in Morris water maze assessments. (AD) Ten months male ScKO and protect watch over mice habits in T maze. (AE) Ten months male ScKO and protect watch over mice habits in Y maze. (AF, AG) Electrophysiological recording from ScKO and protect watch over mice. Representative total-cell recordings on SF-1 neurons within the hypothalamus of ScKO and protect watch over mice are shown on panel AF. Quantitation of their mEPSC and mIPSC frequency and amplitude are showed in panel AG (n ≥ 20 cells from 3 mice). Mouse quantity mature in measuring energy expenditure by launch circuit oblique calorimetry: ScKO mice: n = 16 mice; Alter mice: n = 16 mice. Mouse quantity mature in habits assessments: Alter: n = 24 mice, ScKO mice: n = 17 mice. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001, Kaplan–Meier survival estimate for survival curve. Unpaired t test for behavioral statistics. Statistical capabilities between groups across a couple of time aspects were analyzed by repeated-measures ANOVA. Diversified statistical capabilities were analyzed by one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 1 can be show in S1 Info. mEPSC, little excitatory postsynaptic new; mIPSC, little inhibitory postsynaptic new; SF-1, steroidogenic component-1.
SF1 furthermore expresses within the adrenal glands, pituitary, sexual gland, and spleen [20,21], cre-mediated recombination and deletion of Menin may be “eyombi in these tissue in SF1 Cre Men1f/f mice. We indeed discovered that the Menin levels were diminished in these glands (S4 Fig). To exclude the outcomes of Menin deletion in varied glands, we injected cre recombinase-dependent virus (AAV-CAG-Cre) into the VMH of Men1f/f and WT mice to namely delete Menin in VMH situation (Fig 2A and 2B). Menin knockdown in neurons of VMH of AAV-CAG-Cre-Men1f/f mice without principal toxicity that outcomes neuronal demise were confirmed by western blotting, proper time-PCR, and immunostaining (Figs 2C–2F, S5E, S5F, and S5H–S5K). AAV-CAG-Cre-Men1f/f mice furthermore acquire a diminished lifespan without principal adjustments in physique weight, brain weight, and neuron quantity in hypothalamus and hippocampus when put next with AAV-CAG-Cre–injected protect watch over mice (S5A–S5D and S5G Fig). We furthermore conducted launch circuit oblique calorimetry, the hypothalamic inflammation stage assessments, cognitive behavioral experiments, and discovered that AAV-CAG-Cre-Men1f/f mice showed same outcomes to ScKO mice (Fig 2G–2R). These outcomes counsel that Menin deficiency within the hypothalamus bolt up rising old, doubtlessly thru the improved hypothalamic inflammation.
Fig 2. Menin knockdown virus in VMH outcomes in deficient spatial memory and metabolic dysfunction.
(A) Schematic diagram of knockdown of Men1 by injection of Cre-dependent CAG-GFP-AAV in VMH of Men1f/f mice and protect watch over mice (injection at 9M) that are defined as AAV-CAG-Cre and AV-CAG-Cre-Men1f/f. (B) AAV-GFP immunofluorescence staining in mouse hypothalamus following AAV injection. Scale bar, 100 μm. (C) Representative hypothalamus brain sections from 10 months male AAV-CAG-Cre–injected WT mice and AAV-CAG-Cre–injected Men1f/f mice stained with Menin (crimson) and GFP (inexperienced) antibody. Piece used to be counterstained with DAPI (blue). Scale bar, 100 μm, 20 μm. (D–F) The protein and mRNA levels of Menin within the hypothalamus from 10 months feeble AAV-CAG-Cre–injected WT and Men1f/f mice; n = 3 mice. (G–Okay) Morris water maze (G–I), T maze (J) and Y maze (Okay) assessments were completed in 10 months male AAV-CAG-Cre–injected WT and Men1f/f mice (AAV-CAG-Cre/AAV-CAG-Cre-Men1f/f). All thru Morris water maze assessments, 10 months male AAV-CAG-Cre–injected WT and Men1f/f mice were analyzed for secure away latency at some stage in a 6-day coaching duration (G). On the next day, mice were analyzed for time spent within the map zone and varied quadrants (northeast, southeast, and northwest) (H), resolution of map crossings and time required from entrance to the map platform (I). (L) Day and evening respiratory quotients (RER) of 10 months male AAV-CAG-Cre mice and age-matched AAV-CAG-Cre-Men1f/f mice were measured. Loyal dietary (M), water intake (N), and exercise (O) were furthermore measured. (P–R) Inflammatory components protein expression and mRNA levels within the hypothalamus of 10 months male AAV-CAG-Cre mice and AAV-CAG-Cre-Men1f/f mice; n = 3 mice. (S–U) DEGs were identified from the hypothalamus of 13 months male ScKO mice and protect watch over mice and were shown in panel S. DEGs then were analyzed by GO pathway (T) and KEGG pathway (U). The crimson field represents the enriched metabolic pathway. The blue field represents the enriched cytokine pathway; n = 3 mice. Mouse quantity mature in measuring energy expenditure by launch circuit oblique calorimetry: AAV-CAG-Cre mice: n = 8 mice; AAV-CAG-Cre-Men1f/f mice: n = 8 mice. Mouse quantity mature in habits assessments: AAV-CAG-Cre mice: n = 21 mice; AAV-CAG-Cre-Men1f/f mice: n = 22 mice. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001. Unpaired t test for behavioral statistics. Statistical capabilities between groups across a couple of time aspects were analyzed by repeated-measures ANOVA. Diversified statistical capabilities were analyzed by one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 2 can be show in S2 Info. DEG, differentially expressed gene; VMH, ventromedial hypothalamus.
Menin deficiency outcomes in disrupted metabolism within the hypothalamus
To secure a deeper insight into how Men1 deletion in VMH regulates systematic rising old, we then completed next-generation sequencing to title differentially expressed genes (DEGs) in VMH of 13-month-feeble protect watch over and ScKO mice (S2 Fig and S1 Dataset). The diagnosis revealed a total of 681 genes with differential expression, including Acox2, B3gnt5, Ugtla6a, and Pklr that are obsessed on metabolic pathway (Fig 2T and 2U).
To make clear detailed adjustments in metabolomics, we screened 40 fundamental metabolites (S6 Fig) and completed METARECON [22,23] diagnosis. The METARECON map, which contains the covariance records matrix of the measured metabolite profiles from VMH of ScKO and protect watch over mice alongside side Eq 1, used to be mature to title metabolic perturbation aspects. Basically the most attention-grabbing perturbation within the differential Jacobian, when evaluating VMH of ScKO versus controls, mixed with the centered metabolomics, used to be detected for the response payment elasticity of NADH to KEG (ðf NADH/ðf KEG) (Fig 3C), which the adjustments in D-serine synthesis pathway (Fig 3A–3D) used to be highlighted in ScKO mice. Moreover, reduction in D-serine levels were show in serum of feeble folks (Fig 3E).
Fig 3. Menin deficiency outcomes in disrupted metabolism within the hypothalamus.
(A) PCA of the metabolomics records revealing an glaring separation between the hypothalamus of 13-month-feeble ScKO mice and protect watch over mice. (B) For inverse modeling of biochemical regulation from metabolomics covariance records, a metabolic reconstruction and pathway reduction from metabonomics is completed (RECON). The metabolite variance of two groups is visible, which is extra exploited for the calculation of the Covariance matrix (COV) and attributable to this truth for the Jacobian matrix (JAC) the spend of the stochastic Lyapunov matrix Eq 1 (look the METARECON map in Materials and methods). (C) Differential Jacobian matrix of the hypothalamus of 13-month-feeble ScKO mice and protect watch over mice derived from covariance records from the metabolomics datasets. All entries represent median values of 103 calculations normalized to the sq. of interquartile distance. dF and dM represent the entries of the Jacobian matrix and consult with Eq 2 (look the METARECON map in Materials and methods). Basically the most attention-grabbing perturbation used to be identified because the Jacobian entry ðfNADH = ðfKGA, pointing to D-serine synthesis pathway within the underlying biochemical community in panel D. (D) Simplified biochemical interplay community of D-serine synthesis pathway adjusted to the measured metabolites. (E) D-serine levels in serum from young (22-26Y) and feeble folks (83-94Y) were clear by ELISA; n = 5. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001. Statistical capabilities were analyzed by one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 3 can be show in S3 Info. PCA, major component diagnosis.
De novo D-serine synthesis is catalyzed by phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase (PSAT1), phosphoserine phosphatase (PSPH) [24], and serine racemase (SRR) (Fig 4A). It starts from conversion of 3-phosphoglycerate into 3-phosphohydroxypyruvate, catalyzed by PHGDH, the first payment-limiting enzyme of serine synthesis. Cleave value in D-serine levels were extra confirmed by measuring D-serine levels within the hypothalamus of ScKO mice and controls (Fig 4B), as successfully as within the hypothalamus of AAV-CAG-Cre-Men1f/f and controls (Fig 4C). We then discovered that the expression of PHGDH, the first payment-limiting enzyme of serine synthesis, diminished a good deal in VMH of AAV-CAG-Cre-Men1f/f (Fig 4D–4H) and ScKO mice (S7 Fig) when put next with controls. These records urged that the diminished D-serine in Menin deletion situation is induced by the decline of PHGDH.
We next explored the mechanism underlying how Menin regulates expression of PHGDH. Menin contributes to epigenomic modulation of gene expression, partly thru its association with H3K4me3 modification [25]. To verify whether or no longer Menin regulates PHGDH thru H3K4me3, we completed H3K4me3 chromatin immunoprecipitation (ChIP) assays by 5 clear primer pairs focusing on the phgdh promoter locus (Fig 4I) and discovered sturdy H3K4me3 binding within the phgdh promoter situation from −1978 to −123 in major neurons (Fig 4J). Moreover, Menin-ChIP assays demonstrated Menin occupancy of the phgdh promoter situation (Fig 4K). The ChIP PCR bands from above assessments can be show in S8 Fig. We then overexpressed Menin in VMH by injecting Menin-AAV in 20-month-feeble mice (Fig 4L) and discovered the expression of PHGDH were a good deal increased (Fig 4M and 4N). Extra importantly, the levels of D-serine within the hypothalamus of extinct mice were furthermore increased after overexpression of Menin (Fig 4O). These outcomes counsel that Menin binds to phgdh promoter situation and facilitates chromatin transforming for phgdh transcription, which regulates D-serine synthesis.
Fig 4. Menin deficiency outcomes in disrupted D-serine synthesis within the hypothalamus.
(A) Schematic diagram of D-serine synthesis pathway. (B, C) D-serine levels in lysates of the hypothalamus from 10 months male protect watch over and ScKO mice (B), and 10 months male AAV-CAG-Cre and AAV-CAG-Cre-Men1f/f mice (C) were clear by ELISA; n = 6 mice. (D–F) The protein and mRNA levels of PHGDH within the hypothalamus from 10 months male AAV-CAG-Cre and AAV-CAG-Cre-Men1f/f mice were measured; n = 3 mice or n = 6 mice, respectively. (G, H) Immunofluorescent staining of PHGDH (crimson) and GFP (inexperienced) within the hypothalamus situation from 10 months male AAV-CAG-Cre and AAV-CAG-Cre-Men1f/f mice. Representative confocal photos are shown on panel G. Scale bar, 100 μm, 20 μm. Quantitation of fluorescence intensity of PHGDH are showed in panel H; n = 3 mice. (I) Schematic diagram of phgdh promoter situation. (J, Okay) ChIP assays the spend of antibodies against H3K4me3 or Menin were completed in cultured wild-fashion neurons on DIV 12; n = 3 just experiments. (L–N) AAV-Menin-GFP virous or protect watch over virous AAV-GFP were injected into the hypothalamus situation of 20 months male mice (Frail+AAV:GFP; Frail+AAV:Menin-GFP). The protein and mRNA levels of Menin and PHGDH were measured within the hypothalamus of above mice; n = 3 mice. (O) D-Serine levels within the hypothalamus from 20 months male Frail+AAV:GFP and Frail+AAV:Menin-GFP mice were clear by ELISA; n = 4 mice. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001, one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 4 can be show in S4 Info. ChIP, chromatin immunoprecipitation; PHGDH, phosphoglycerate dehydrogenase.
Restoring Menin within the hypothalamus ameliorates rising old
We constructed a Cre recombinase-dependent Menin-AAV with EGFP expression and a CMV-cre AAV with mCherry expression. These 2 AAVs were mixed and bilaterally injected into VMH of 20-month-feeble mice to restore Menin expression in VMH (Fig 5A and 5B). Overall, up-regulating Menin in VMH of 20-month-feeble mice may per chance per chance successfully fashion bigger mouse lifespan (Fig 5C) without principal adjustments in physique weight, brain weight, and neuron quantity in hypothalamus and hippocampus (S9A–S9D Fig). Thirty days after AAV injection, Menin overexpression a good deal reversed systemic rising old phenotypes, including pores and skin thickness, bone mass, and tail tendon collagen immoral-linking (Fig 5D–5G). Seriously, varied rising old-linked phenotypes, reminiscent of inflammation levels, irregular metabolic circadian rhythm, and food intake were furthermore a good deal ameliorated by overexpression of Menin in VMH (Fig 5H–5L). Behavioral assessments were attributable to this truth completed 30 days after AAV injection. Seriously, restoring Menin expression a good deal improved the final behavioral performance, including the impaired studying, cognition, and balance exercise of feeble mice (Fig 5M–5S). Some of the feeble mice had a 0 bag on T maze or Y maze test, suggesting that the low bag may per chance per chance no longer handiest replicate rising old-linked cognitive decline nevertheless furthermore rising old-linked decline of physical distress. To peek this possibility, we measured the free exploration distance in launch discipline assessments and discovered that the commute distance used to be a good deal improved in feeble mice injected with AAV:Menin-GFP (VMH Menin overexpression) than feeble mice with AAV:GFP (protect watch over mice), nevertheless there used to be no principal difference within the swimming bolt within the water maze between the 2 groups (S9E–S9G Fig).
Fig 5. Enhanced Menin expression within the hypothalamus mice extends lifespan and ameliorates rising old-linked phenotype.
(A, B) Detailed schematic diagram of overexpression of Menin by AAV in VMH of feeble male mice (20M). (C) Lifespan of those mice (n = 19 mice per community). (D–G) These mice were sacrificed for measuring dermal thickness (D, E), bone mass (F), and tail tendon breaking time (G); n = 4 mice. Scale bar, 100 μm. (H, I) The day and evening respiratory quotients (RER) (H) and feed intakes (I) of 20 months male Frail+AAV:GFP and Frail+AAV:Menin-GFP mice were measured. (J–L) Inflammatory components protein expression and mRNA levels within the hypothalamus of 20 months male Frail+AAV:GFP and Frail+AAV:Menin-GFP mice; n = 3 mice. (M–S) Behavioral diagnosis of 20 months male Frail+AAV:GFP and Frail+AAV:Menin-GFP mice by Morris water maze assessments (M–O), Y maze (P), T maze (Q), launch discipline (R), and rotarod test (S). Mouse quantity mature in measuring energy expenditure by launch circuit oblique calorimetry: Frail+ AAV:GFP: n = 8 mice, Frail+AAV:Menin-GFP: n = 8 mice. Mouse quantity mature in habits assessments: Frail+AAV:GFP: n = 14 mice, Frail+AAV:Menin-GFP: n = 16 mice. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001, Kaplan–Meier survival estimate for survival curve. Unpaired t test for behavioral statistics. Statistical capabilities between groups across a couple of time aspects were analyzed by repeated-measures ANOVA. Diversified statistical capabilities were analyzed by one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 5 can be show in S5 Info. VMH, ventromedial hypothalamus.
D-Serine complement reduces cognitive decline in ScKO mice
ScKO mice exhibited impaired cognition suggests that the neuronal aim in hippocampus may per chance per chance has been dysregulated. We first examined the VMH-hippocampus neural projections and discovered there’s no projection difference between protect watch over and ScKO mice (Fig 6A–6C). However the D-serine levels were a good deal diminished in hippocampus of ScKO mice versus controls (Fig 6D), as successfully as in hippocampus of AAV-CAG-Cre-Men1f/f versus controls (Fig 6E). Meanwhile overexpression of Menin in VMH a good deal increased hippocampal D-serine levels in 20 months feeble mice (Fig 6F). Knockdown of Menin in VMH furthermore lowered PSD95 and Synaptophysin expression in hippocampus (Fig 6G–6J). Golgi staining extra signifies that the dendritic density of hippocampal neurons used to be furthermore a good deal lowered (Fig 6K–6M). Next, we completed an electrophysiological characterization of synaptic aim in hippocampal CA1 situation on high of things and ScKO mice. We noticed a of route huge reduction in high-frequency stimulation (HFS)-induced long-term potentiation (LTP) in ScKO mice (Fig 6N and 6O).
Fig 6. VMH Menin knockdown induced diminished hippocampal D-serine levels and impaired synaptic capabilities.
(A) The schematic diagram of virus injection. AAV-CamkIIα-EYFP (200 nL) used to be injected into VMH of ScKO mice and protect watch over mice. Scale bar, 100 μm. (B, C) A representative EYFP-labeled fiber labeled in CA1 neuron. Representative photos are shown in panel C. Scale bar, 20 μm. Quantitation of fluorescence intensity is shown in (C); n = 5 mice. (D–F) D-serine levels in hippocampus from 10 months male protect watch over and ScKO mice lysates (D), 10 months male AAV-CAG-Cre and AAV-CAG-Cre-Men1f/f mice lysates (E), and Menin overexpressed within the hypothalamus situation of feeble mice (F) were clear by ELISA; n = 4 or 6 mice. (G–J) Immunofluorescent staining of synaptophysin (crimson) and PSD95 (inexperienced) in hippocampus from 10 months male protect watch over and ScKO mice (G), and 10 months male AAV-CAG-Cre and AAV-CAG-Cre-Men1f/f mice (H). Representative confocal photos are shown on panel G and H, respectively. Scale bar, 20 μm, 5 μm. Quantitation of resolution of puncta of synaptophysin and PSD95 are showed in panel I and J, respectively, n = 6 slices from 3 mice. (Okay–M) Six months male ScKO mice and protect watch over mice were subjected to Golgi staining. Representative Golgi staining from cortex and hippocampal CA1 regions is shown in panel Okay. Scale bar, 100 μm, 5 μm; n = 3 mice. Quantitation of dendritic complexity in neurons from above mice used to be shown in panel L and M; n = 20 neurons. (N, O) LTP recordings from 6 months male ScKO and protect watch over mouse brain (n = 9 slices from 3 protect watch over mice; 8 slices from 4 ScKO mice). Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001, one-device ANOVA with Tukey’s post hoc diagnosis. The underlying records of Fig 6 can be show in S6 Info. LTP, long-term potentiation; VMH, ventromedial hypothalamus.
We then wondered whether or no longer D-serine complement can attenuate the cognition decline in ScKO mice and feeble mice. 9 months feeble ScKO and protect watch over mice, 22 months feeble mice were given D-serine in water for 3 weeks (approximately 100 mg/kg physique weight) [26] and then subjected to following assessments, respectively (Figs 7A and S10A). The D-serine levels measured by ELISA indeed increased in serum, the hypothalamus and hippocampus of mice (Figs 7B–7D and S10B). Behavioral assessments revealed that D-serine complement rescued the cognitive deficits of 10-month-feeble ScKO mice or 22-month-feeble mice (Figs 7E–7I and S10E–S10I). The D-serine complement furthermore reversed metabolic complications in ScKO mice (Fig 7J–7L) and hippocampus PSD95 and Synaptophysin expression levels in ScKO mice and feeble mice (Figs 7M and 7N, S10J, and S10Okay). These outcomes counsel that D-serine reduction play a actually critical aim within the cognition decline, and its complement can attenuate the cognition decline in ScKO mice and feeble mice.
Fig 7. D-Serine complement reduces cognitive decline in ScKO mice.
(A) Schematic diagram of chronic oral D-serine supplementation (drinking water for 3 weeks). (B–D) D-serine levels in serum and lysates of hypothalamus and hippocampus from 10 months male protect watch over mice, protect watch over +D-serine mice, ScKO mice and ScKO+D-serine mice were clear by ELISA; n = 4 mice. (E–I) Behavioral assessments of the above mice by Y maze (E), T maze (F), and Morris water maze assessments (G–I). (J–L) The above mice were subjected to measure energy expenditure by launch circuit oblique calorimetry. Day and evening respiratory quotients (J), Loyal dietary (Okay), and feed/water intake (L) were measured. (M, N) Immunofluorescent staining of synaptophysin (crimson) and PSD95 (inexperienced) in hippocampus from 10 months male protect watch over mice, ScKO mice, and ScKO+D-serine mice. Representative confocal photos are shown on panel N. Scale bar, 20 μm, 5 μm. Quantitation of resolution of puncta of synaptophysin and PSD95 are showed in panel O, n = 6 slices from 3 mice. Mouse quantity mature in habits assessments: Alter: n = 12 mice, Alter+ D-serine supplementation: n = 16 mice, ScKO: n = 15 mice, ScKO+ D-serine supplementation: n = 18 mice. Mouse quantity mature in measuring energy expenditure by launch circuit oblique calorimetry: Alter: n = 8 mice, Alter+ D-serine supplementation: n = 8 mice, ScKO: n = 8 mice, ScKO+ D-serine supplementation: n = 8 mice. Knowledge represent mean ± SEM, n.s.: no longer principal, *p < 0.05, p < 0.01, p < 0.001, p < 0.0001. Unpaired t test for behavioral statistics. Statistical applications between groups across multiple time points were analyzed by repeated-measures ANOVA. Other statistical applications were analyzed by one-way ANOVA with Tukey’s post hoc analysis. The underlying data of Fig 7 can be found in S7 Information.
Discussion
The present study reported the important function of VMH Menin in regulating systemic aging. Manipulating Menin levels selectively in SF-1 neuron of VMH altered a variety of aging biomarkers in multiple systems and the aging process of whole body, through regulation of hypothalamic microinflammation and metabolic states (e.g., serine signaling). These results indicate Menin as a novel arbiter for systemic aging and the therapeutic potential for D-serine in treatment against cognitive decline.
The microinflammation of the hypothalamus leads to aging, which can be reversed by GnRH secreted by the pituitary gland [5]. The metabolic abnormalities of the body can also lead to microinflammation of the hypothalamus [8,9]. Menin is a powerful epigenetic regulation of gene transcription and cell signaling [27], affecting the secretion of multiple glands on the HPA axis and the metabolism of various organs in the body. At the same time, according to our previous findings [14], Menin could bind to the promoter region of p65 and inhibited its transcription, and the lost function variant of MEN1 (SNP rs375804228) failed to inhibit p65 transcription [14]. It is plausible that decreased Menin signaling contribute to the activated neuroinflammation in the hypothalamus.
In this article, we found that the expression of hypothalamic Menin decreases with age. The hypothalamic deficiency of Menin leads to an accelerated aging process, while the overexpression of Menin reverses systematic aging phenotype. Therefore, we speculate that the decline of Menin expression in the hypothalamus with age may be one of the driving factors of aging and Menin may be the key protein connecting the genetic, inflammatory, and metabolic factors of aging.
Metabonomics analysis and METARECON strategy indicated that D-serine biosynthesis pathway was significantly altered with Menin deficiency, suggesting that de novo D-serine biosynthesis in SF-1 neuron in VMH depends on Menin signaling. Previous research found that Menin is closely associated with glycometabolism and Menin inhibitors induced increase in glycolysis occurs in an mTOR-independent manner, which enhances the sensitivity of colorectal cancer cells to EGFR inhibitors [28]. Another study found that Menin regulates the D-serine biosynthetic pathway in Ewing sarcoma by inhibiting phgdh transcription [29], which is consistent with the findings in present study.
Previous studies identified that VMH SF-1 neurons act as important metabolic regulators [15–17] and play an important role in maintaining whole-body energy homeostasis. L-Serine is synthesized from 3-phosphohydroxypyruvate by a serial of reactions mediated by PSAT1 and PSPH [24,30]. L-serine is further converted to D-serine by D-serine synthase serine racemase (SRR). The main target of D-serine is N-methyl-D-aspartate receptor (NMDAR), which is essential for neurotransmission, learning, and memory formation, especially in CA1 and dentate gyrus of hippocampus [31–35]. Although several studies report that this L-serine/D-serine transformation also exit in astrocyte, recent new findings demonstrate that serine racemase is predominantly expressed by neuronal structures, suggesting that a significant fraction of D-serine comes from neurons [36–39]. The D-serine level is significantly decreased in conditional neuronal SRR-KO mice, suggesting the neuronal D-serine source. Interestingly, these mice also exhibit hippocampal LTP deficits and reduced NMDAR synaptic potentials [37,38]. On the other hand, conditional astrocytic SRR-KO mice display only a marginal decrease in brain serine racemase expression, with no changes in brain D-serine or NMDAR synaptic potentials [37,38]. These data suggested that serine racemase predominantly express in neurons [36,37,39]. Our data also suggested that D-serine may be a neuronal transmitter that is secreted by SF1 neurons.
Our data indicates that the remission of aging phenotype by D-serine is limited to cognitive improvement, leaving peripheral systems aging phenotypes unchanged. This may be due to the limited time of D-serine supplement, or there are other downstream pathways regulated by Menin that contributes to peripheral aging regulation.
By co-immunostaining Menin with SF-1, GFAP, and IBA1, we found that, with the increase of age, the expression of Menin decreased significantly only in SF-1 neurons, but not in astrocytes and microglia (S1M–S1R Fig). Therefore, the anti-aging effect of Menin in VMH is mainly mediated neuronal Menin. Hypothalamic Menin signaling diminished in neuron leads to increased levels of inflammation. This probably because the deficiency of Menin in neurons affects the interaction between neurons and glial cells in a certain way, and then leads to the release of cytokines from glial cells. We previously found that Menin can associate with p65 to inhibit NF-κB transactivation [14], inhibiting the neuroinflammation in astrocyte. Glial Menin may also play a role in inhibiting inflammation in VMH, which needs take the study forward in the further. Whether the up-regulation of cytokines is connected to D-serine biosynthesis also needs further studies.
By SF1-Cre mice, we deleted Menin at early stage of mice. The ScKO mice exhibited a premature aging phenotype in the middle of the age. To avoid the developmental adaptations of ScKO mice, we used viral-Cre approach to knockdown Menin in adult mice that also showed aging phenotype. Since the expression of Menin gradually decreases with age, earlier and longer knockout of Menin will lead aging appear earlier and severer.
For visualization the subcellular localization of Menin, we attached GFP with Menin when constructed AAV virus. The GFP fused Menin can up-regulate the transcription of phgdh and improve the synthesis of D-serine compared with GFP control. This data indicates that GFP-Menin functions as Menin alone. We are aware of the potential side effect of GFP fused protein expression, and the inclusion GFP may have potential effect on the normal expression, trafficking, and overall biological function in other ways. However, our data about GFP-Menin seems have no side effects.
Currently, the upstream regulators for Menin are unclear. Previous study showed that GLP-1 signaling-activated protein kinase A (PKA) directly phosphorylates Menin at the serine 487 residue, relieving Menin-mediated suppression of insulin expression and cell proliferation, while somatostatin stimulates Menin by suppression PKA [40,41].
In summary, we have identified the importance of VMH Menin in orchestration of aging pace. Menin levels may indicate the aging status and serve as anti-aging target.
Materials and methods
Human samples
Serum of different age groups of human subjects were acquired from the Department of Health Examination, the first affiliated hospital of Xiamen University. Study protocols received prior approval from the Ethics Committee of the first affiliated hospital of Xiamen University. The approval number of the protocol is XDYX202101K06. Informed content was written and obtained from all subjects. The samples were collected in adherence with the Declaration of Helsinki. Characteristic information can be found in Table 1.
Animals
All mice were maintained within the laboratory animal center at Xiamen University, and all experimental procedures involved were performed according to protocols approved by the Institutional Animal Care and Use Committee at Xiamen University. The approval number of the protocol is XMULAC20200054. We also abide by the provisions of the Biosafety Law of the People’s Republic of China, the Regulations on the Administration of Experimental Animals, the National Standards for Experimental Animals (GB14925-2010), the Guidelines for Ethical Review of the Welfare of Experimental Animals (GBT 35892–2018), and the relevant rules and regulations formulated by Xiamen University. Mice were housed under a 12 h light/dark cycle with free access to standard rodent chow and water. Each cage housed a maximum of 4 mice. Mice were maintained under specific-pathogen-free (SPF) conditions and were not subject to immune suppression. Health of the animals used was regularly controlled by animal caretakers. All mice used were drug/test naïve. Six months male and female ScKO mice and control mice were used to test body weight, brain weight, Menin levels in CNS and peripheral tissues, and the hypothalamic inflammation levels. Six months male ScKO mice and control mice were used for Golgi staining and LTP recording. Male ScKO mice and control mice, male Old+AAV:GFP mice and Old+AAV:Menin-GFP mice were used to record lifespan. Ten months male and female ScKO mice and control mice were employed in behaviors tests. Ten months male ScKO mice and control mice were employed in open circuit indirect calorimetry, muscle fiber size, bone mass, skin thickness, myocardial thickness, β-Gal stain, and tail tendon collagen cross-linking tests. The hypothalamus of 13 months male ScKO and control mice were used for the RNA-seq tests and targeted metabolomics. Ten months male AAV-CAG-Cre mice and AAV-CAG Cre-Men1f/f mice were employed in behaviors tests and open circuit indirect calorimetry. Twenty months male Old+AAV:GFP mice and Old+AAV:Menin-GFP mice were employed in behaviors tests, open circuit indirect calorimetry, bone mass, skin thickness, and tail tendon collagen cross-linking. Animals were used according to “3Rs” principles (Replacement, Reduction, and Refinement) in all experimental procedures.
The floxed Men1 mouse strain (Men1f/f) was obtained from Dr. Guanghui Jin and Dr. Xianxin Hua [42]. SF1-Cre transgenic mice were bought from Jackson Laboratory. Men1-ScKO mice were obtained by crossing Men1 floxed mice with respective SF1-Cre mouse lines. Men1 floxed mice were used as controls.
Experimental design
All experiments described in this study were performed a minimum of 3 biological reduplication. We did not use a statistical method to predetermine the proper sample size. The sample size per experiment was determined according to previous publications.
Open circuit indirect calorimetry
The optimum indirect calorimetry system (Sable Promethion) was used for simultaneous measurements of mice undergoing dietary studies at same time. Systems of up to 16 cages were available for such simultaneous measurements. The mice were reared in single cage for 2 days before recording. After that, the mice were put into the calorimetry system to record the energy metabolism of a week. The day-night shift occur in the animal house were 08: 00 and 20: 00. VO2, VCO2, energy expenditure, respiratory quotient, mass of food consumed, mass of water consumed, body weight, and distance in locomotion were recorded.
ChIP
ChIP procedures were performed according to the manufacturer’s instructions (Millipore, 17–295), which have been detailed descripted in the method of our previous study [25].
Generation of AAVs and stereotaxic injection
pAAV-CAG-cre-GFP (virus titer: 5.23 × 1012/mL) were purchased from BrainVTA (Wuhan, China). AAV-CMV-Cre-mCherry (virus titer: 5.02 × 1012/mL) were purchased from OBiO Technology Corperation (Shanghai, China). AAV-EF1α-DIO-MEN1-EGFP-WPRE (virus titer: 5.05 × 1012/mL) and AAV-EF1α-DIO-EGFP-WPRE (virus titer: 5.62 × 1012/mL) were purchased from BrainVTA (Wuhan, China). We attached GFP with Menin in a classical fusion to visualize the subcellular overexpression of Menin when constructed AAV virus. Packaged viruses were stereotactically injected into the VMH of control mice or men1f/f mice respectively as described previously. All mice were anesthetized and placed in a stereotaxic frame, a skin incision was made and holes were drilled at VMH (x(±0.4 mm from bregma) and y(−1.58 mm from bregma)). A total of 0.2 μl viruses were delivered at 0.20 μl/min at z-depths of 5.7 mm. The syringe was left in place for 10 min after each injection, before being withdrawn slowly. rAAV-CaMKIIα-EYFP-WPRE-pA (virus titer: 3.26 × 1012/mL) were purchased from Brain VTA (Wuhan, China). To investigate the projection from the VMH toward to the hippocampus, anterograde virus tracer AAV CaMKIIα-EYFP was stereotactically injected into the VMH in ScKO and control mice, respectively. The mice were allowed to survive for 3 weeks and then to observe EYFP-labeled fibers within the hippocampus.
Electrophysiology
For hippocampal slice LTP recordings procedure, please refer to the method of our previous study [14].
Whole-cell patch clamp recordings were obtained from SF-1 neurons in the hypothalamus of ScKO mice and control mice using 4 to 8 MΩ borosilicate glass pipettes (Harvard Apparatus). The pipette recording solution contained (in mmol 1−1): 8.0 NaCl, nominally 0.0001 CaCl2, 0.3 Na-GTP, 130 potassium gluconate, 10.0 Na-Hepes, 1.0 MgCl2, and 2.0 Na-ATP (pH adjusted to 7.4 with methanesulfonic acid; 295 to 300 mosmol 1−1). The pipettes with an Ag–AgCl electrode were connected to a CV-4 head stage and an Axopatch-1D amplifier with a Digi data 1200 interface (Axon Instruments). The pipettes are positioned within the tissue using a motorized patch-clamp micromanipulator. Seal resistance was typically 4 to 10 GΩ. Typical whole-cell access resistance (Ra) was 5 to 30 MΩ and whole-cell leak was below 20 pA [43]. Miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) were recorded in the presence of tetrodotoxin (500 nmol/L). We recorded mEPSCs and mIPSCs at holding potentials of –70 and 0 mV, respectively, in the same cell (3 min each; n > 20 cells/community).
Supplementation of D-serine
ScKO and protect watch over mice were given D-serine (600 mg/L, Sigma-Aldrich, St. Louis, Missouri, United States of The US) within the drinking water for 3 weeks. Every mouse took about 5 mL per day.
RNA-sequencing diagnosis
The hypothalamus of ScKO and protect watch over mice were harvested. Remoted RNA used to be got for RNA-sequencing diagnosis [14]. cDNA library constructing and sequencing were completed on Illumina platform by Beijing Novogene Corporation. High of the variety reads were aligned to the mouse reference genome the spend of Bowtie 2. Expression levels for every of the genes were normalized to fragments per kilobase of exon model per million mapped reads (FPKM) the spend of RNA-seq by Expectation Maximization (RSEM). Genes with ≥2-fold alternate and p < 0.05 were belief about to be statistically principal. DEGs between samples were identified and clustering diagnosis. Then, purposeful annotations were completed.
ELISA
D-Serine levels in mouse serum, hippocampus, and hypothalamus were measured the spend of ELISA Assay Kits (D-serine Elisa Equipment, CAT CZY-041905M, Nanjing Caobenyuan Biotech Co.) in step with the manufacturer’s instructions.
Basic neuron culture procedures
Basic neurons were dissected from timed-pregnant females at E16.5. Rapid, brain cortices were dissected from pups of varying genotypes. The Meninges were eradicated, and cortical tissue used to be dissociated by enzymatic digestion. Remoted major neurons were plated on poly-D-lysine–coated dishes and cultured in Neurobasal medium supplemented with B27 (Gibco)/1% penicillin/streptomycin (Invitrogen) and maintained in a 5% CO2 incubator at 37°C.
World DNA methylation assay
DNA methylation levels in mouse hypothalamus were measured the spend of World DNA Methylation Assay Equipment (World DNA Methylation Assay Equipment, ab233486, Abcam) in step with the manufacturer’s instructions.
Senescence-associated β-galactosidase assay with immunostaining
Senescence β-galactosidase staining used to be completed the spend of a colorimetric package (Abcam) in step with the manufacturer’s protocol.
Behavioral be taught
For your entire behavioral assessments procedures (T and Y maze, forced swimming test, tail suspension test, sucrose preference test (SPT), sucrose consumption test (SCT), rotarod test, launch discipline test, and Morris water maze), please consult with the programs in our old eye [14].
Western blotting
Cultured cells and mouse brain tissues (encompass hypothalamus and hippocampus) were homogenized in lysis buffer (RIPA) on ice for 40 min and attributable to this truth centrifuged at 12,000 rpm for 10 min at 4°C. Supernatants were transferred to a clear 1.5 mL tube, and protein samples were resolved by SDS-PAGE (sodium dodecyl sulfate–polyacrylamide gel electrophoresis) and subsequent immunoblotted onto polyvinylidene difluoride (PVDF) membranes. Sample containing 30 μg of protein used to be separated the spend of 10% SDS-PAGE gels. Proteins were transferred onto PVDF membranes in an ice-cool buffer (25 mM Tris-HCl, 192 mM glycine, and 20% methanol) by electro transfer for 1.5 h. Immunoblots were probed with indicated antibodies. Goat-anti-mouse secondary antibodies and goat-anti-rabbit secondary antibody were purchased from Millipore (#AP132P, #AP124P). Quantification of band intensities were normalized to β-actin, and averaged from on the least 3 just experiments.
Immunofluorescence
Mouse brain sections or cultured cells were washed thrice with PBS and antigen retrieval used to be completed the spend of citrate buffer (pH 7.0); samples were then permeabilized and blocked in PBS containing 0.5% Triton X-100 and 10% stylish goat serum at room temperature for 1 h. Sections were incubated with major antibodies in blocking off buffer overnight at 4°C. After washing, secondary antibodies were added to the blocking off buffer and incubated for 1 h. Samples were then washed and counterstained with DAPI. Shots were obtained the spend of a Nikon confocal microscope.
Basic antibodies mature for immunostaining encompass: Menin (rabbit, 1:1,000; Abcam, Ab4452; mouse, 1:1,000; Thermo Fisher, A500-0003A), PHGDH (rabbit, 1: 300; Life Span, #LS-C81937-50), PSAT1 (rabbit, 1: 100; Life Span, #LS-C80875-50), PSPH (rabbit, 1: 100; lifespan, #LS-C38237-50), SF1(rabbit,1: 500; Abcam, Ab168380), GFAP (mouse,1: 500; Cell Signaling Abilities, #3670), NeuN (mouse, 1:1,000; Millipore, #MAB377), Iba1 (goat, 1: 200; Abcam, Ab48004), 488/594 donkey anti-mouse/rabbit secondary antibodies (1: 500), and Mounting Medium with DAPI used to be purchased from Invitrogen.
Quantitative RT-PCR
Entire RNA from animal tissues and cells were isolated the spend of Trizol reagent in step with the manufacturer’s instructions (Invitrogen). Reverse transcription used to be completed the spend of ReverTra Ace qPCR RT Grasp Mix (Toyobo, FSQ-201). RNA concentrations were adjusted to 2 g/L in nuclease-free water, and total RNA used to be reverse-transcribed in a 20 μL response volume. cDNA used to be amplified by proper-time quantitative RT-PCR the spend of SYBR Inexperienced (Roche) reagent. Samples were assayed in triplicate and actin used to be mature as an interior protect watch over. Primer sequences mature in this eye can be show in Table 2.
Focused metabolomics
Sample preparation.
The hypothalamus of ScKO and protect watch over mice were weighed sooner than the extraction of metabolites and dried lyophilized were floor in a 2 mL Eppendorf tube containing a 5-mm tungsten bead for 1 min at 65 Hz in a Grinding Mill. Metabolites were extracted the spend of 1 mL precooled combinations of methanol, acetonitrile, and water (v/v/v, 2:2:1) and then positioned for 1 h ultrasonic shaking in ice baths. Subsequently, the aggregate used to be positioned at −20°C for 1 h and centrifuged at 14,000 g for 20 min at 4°C. The supernatants were recovered and concentrated to dryness in vacuum.
UHPLC-MS diagnosis.
The LC/MS fragment of the platform used to be in step with a Thermo Fisher Vanquish UHPLC equipped with an ACQUITY UPLC BEH Amide column (1.7 μm, 2.1 mm × 100 mm, Wasters) and a Thermo-TSQ Vantage mass spectrometer. Energy metabolites were monitored in electrospray adversarial-ionization and sure-ionization mode. The 2 μL samples were injected sequentially into a Thermo-TSQ Vantage mass spectrometer equipped with a Vanquish UHPLC system with autosampler (Thermo Fisher). The ACQUITY UPLC BEH Amide column (1.7 μm, 2.1 mm × 100 mm, Wasters) used to be heated to 45°C below a drift payment of 300 μL/min. A gradient used to be mature to separate the compounds consisted of 20 mM ammonium acetate (solvent A) and 5% acetonitrile (solvent B). The gradient started at 5% solvent A for 1 min and rising linearly to 35% solvent A over 13 min, and then rising linearly to 60% solvent A over 2 min with a 2-min protect sooner than returning the starting combination at some stage in 0.1 min and re-equilibrating for 4 min. QC samples were injected every 6 or 8 samples at some stage in acquisition.
The MS circumstances were as follows: Collision Gas Tension (mTorr): 1.0; Q1 Height Width (FWHM): 0.70; Q3 Height Width (FWHM): 0.70; Cycle Time (s): 1.500; Capillary Temperature: 350.0°C; Vaporizer Temperature: 350.0°C; Sheath Gas Tension: 35.0; Aux Valve Trot alongside with the circulation: 10.0; Spray Voltage: Optimistic polarity −3,500.0V; Detrimental polarity −3,000.0V; scan fashion: selected response monitoring/a couple of response monitoring (SRM/MRM).
Knowledge preprocessing and filtering.
Raw MRM records info were processed by high discovering, alignment, and filtering the spend of Xcalibur Qual browser utility.
Multivariate statistical diagnosis.
Simcap 14 utility (Umetrics, Umeå, Sweden) used to be mature for all multivariate records analyses and modeling. Knowledge were mean-centered the spend of Pareto scaling. Devices were constructed on major component diagnosis (PCA), orthogonal partial least-sq. discriminant diagnosis (OPLS-DA), and partial least-sq. discriminant diagnosis (PLS-DA). All of the items evaluated were tested for over fitting with programs of permutation assessments. The descriptive performance of the items used to be resolute by R2X (cumulative) (very most attention-grabbing model: R2X (cum) = 1) and R2Y (cumulative) (very most attention-grabbing model: R2Y (cum) = 1) values while their prediction performance used to be measured by Q2 (cumulative) (very most attention-grabbing model: Q2 (cum) = 1) and a permutation test (n = 200). The permuted model must always no longer be ready to foretell classes: R2 and Q2 values on the Y-axis intercept must always be decrease than those of Q2 and the R2 of the non-permuted model. OPLS-DA allowed the determination of discriminating metabolites the spend of the variable importance on projection (VIP). The VIP bag price signifies the contribution of a variable to the discrimination between your entire classes of samples. Mathematically, these ratings are calculated for every variable as a weighted sum of squares of PLS weights. The mean VIP price is one, and in overall VIP values over one are belief about as principal. A high bag is in agreement with a solid discriminatory capability and thus constitutes a criterion for the sequence of biomarkers.
The discriminating metabolites were got the spend of a statistically principal threshold of variable affect on projection (VIP) values got from the OPLS-DA model and two-tailed Pupil’s t test (p price) on the normalized uncooked records at univariate diagnosis stage. The p price used to be calculated by one-device diagnosis of variance (ANOVA) for multi-community diagnosis. Metabolites with VIP values increased than 1.0 and p price no longer as a lot as 0.05 were deemed to be statistically principal metabolites. Fold alternate used to be calculated because the logarithm of the stylish mass response (home) ratio between 2 arbitrary classes. On the more than a few side, the identified differential metabolites were mature to secure heatmap analyses with R package.
METARECON map
To title a particular metabolic pathway which struggling from Menin deletion in VMH, we be aware a unique device termed METARECON. The purposeful integration of GC-MS metabolomics records into a biochemical metabolic community constructing used to be completed by the inverse approximation of the biochemical Jacobian matrix. This approximation right away connects the covariance matrix (COV), which used to be constructed from the experimental metabolomics records, to the metabolic community constructing of the first metabolism. The metabolic community model is equipped in S6A Fig.
In the initiating, an acceptable simulation situation and records constructing are made. The generic form of 1 is is known as the “Lyapunov Equation,” which is widely applied to manipulate programs.
(1)
where JAC represents the Jacobian matrix, the COV matrix is derived from the organic variance of just replication diagnosis between a residence of samples, and FLU is a fluctuation matrix that integrates a Gaussian noise aim simulating metabolic fluctuations below stylish-philosophize circumstances. Jacobian matrix JAC is calculated from diffusion matrix FLU and covariance matrix COV created from metabonomic records; 1 no longer handiest considers the noise within the records captured by the fluctuation matrix, nevertheless furthermore combines the statistical traits of the records with the dynamic traits of a trusty system.
Then, the metabolism pathway efficiency consistent is made. In a biochemical atmosphere, r represents the charges for every response and C represents metabolite concentration adjustments, every component of JAC device the pliancy of response charges to any alternate of metabolite concentrations. The corresponding Jacobian is a matrix of all first-whine partial derivatives of all capabilities ri on all metabolites Cj, as shown in Eq 2. The Jacobian matrix JAC in 1 is defined as
(2)
The following step is to consolidate dynamic equation of organic response. On this algorithm, Jacobian matrix is mature to report the native dynamic traits arrive the stylish philosophize of the system.
A residence of differential equations represent the dynamics of metabolic pathways still of a couple of metabolites. As shown in below, where response charges r(r1, r2,⋯rn) are the capabilities of metabolite concentrations C(C1, C2,⋯Cn) over time.
By this vogue, as described in Eq 4, the matrix describes the alternate of every and every metabolite and its impact on the alternate of varied metabolites.
The METARECON means links the kinetic equation of the metabolic pathway represented by above to the covariance of the relevant metabolite concentration records represented by Eq 4. The above programs are mixed to extra exchange the records into Eq 2. This involves the integration of precursor synthesis, substrate utilization, and energy conversion pathways in organic metabolism.
The final step is model enchancment thru metabolic interplay reannotation. JAC matrix has more just variables than symmetric covariance matrix, so parametric answer is wished to secure rid of the uncertainty. By quoting RENEW to search out out the linear system, the answer of Jacobian matrix is got. RENEW is a metabolic interplay matrix, furthermore known as a stoichiometric matrix, which represents the interdependence of metabolic flux and metabolites. Actually, there exists regulation between metabolites without substances consumption. Then, we are able to resolve the non-zero within the Jacobian matrix by importing the stoichiometric matrix of the metabolic community, and swear capability response within the fundamental biochemical community for the closing diagnosis. Basically based on the above theorem, we give a strict transformation from Eqs 2–5.
Statistical diagnosis
All records provided are expressed as arithmetic mean ± SEM. All statistical analyses were completed the spend of GraphPad Prism version 8.0. Null hypotheses were rejected at p values equal to or increased than 0.05. Kaplan–Meier survival estimate used to be mature for survival curve. We conducted a paired t test on launch circuit calorimetry records of two groups. Unpaired t test used to be applied to 2 groups of behavioral statistics. For statistical comparisons between 2 groups, we first completed a Shapiro–Wilk normality test (Prism) to search out out whether or no longer the records used to be seemingly in overall disbursed. Statistically principal variations between groups were clear the spend of one-device ANOVA. In evaluating a couple of comparisons, Bonferroni programs were mature to adjust p values accordingly to diminish the chance of fashion I errors.
All electrophysiological outcomes were analyzed the spend of Sigma Stat 4 statistical utility. Statistical significance used to be evaluated by one-device ANOVA with Holm–Sidak pair-wise assessments. Values of p < 0.05 were belief about statistically principal. DNASTAR Laser gene utility (version 7.1) used to be mature to compare Sanger sequencing records.
Acknowledgments
We thank Dr. Guanghui Jin and Dr. Xianxin Hua for providing the Men1f/f mice.
References
- 1.
Mitchell SJ, Madrigal-Matute J, Scheibye-Knudsen M, Fang E, Aon M, Gonzalez-Reyes JA, et al. Effects of Intercourse, Tension, and Energy Intake on Hallmarks of Rising old in Mice. Cell Metab. 2016;23(6): 1093–112. Epub 2016/06/16. pmid: 27304509; PubMed Central PMCID: PMC4911707. - 2.
Zhang ZD, Milman S, Lin JR, Wierbowski S, Yu H, Barzilai N, et al. Genetics of low human longevity to info drug discovery for healthy rising old. Nat Metab. 2020;2(8): 663–72. Epub 2020/07/29. pmid: 32719537; PubMed Central PMCID: PMC7912776. - 3.
Zhang W, Qu J, Liu GH, Belmonte JCI. The rising old epigenome and its rejuvenation. Nat Rev Mol Cell Biol. 2020;21(3): 137–50. Epub 2020/02/06. pmid: 32020082. - 4.
Jurk D, Wilson C, Passos JF, Oakley F, Correia-Melo C, Greaves L, et al. Power inflammation induces telomere dysfunction and hastens rising old in mice. Nat Commun. 2014;2: 4172. Epub 2014/06/25. pmid: 24960204; PubMed Central PMCID: PMC4090717. - 5.
Zhang G, Li J, Purkayastha S, Tang Y, Zhang H, Yin Y, et al. Hypothalamic programming of systemic rising old engaging IKK-beta, NF-kappaB and GnRH. Nature. 2013;497(7448): 211–6. Epub 2013/05/03. pmid: 23636330; PubMed Central PMCID: PMC3756938. - 6.
Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, et al. Sirt1 Extends Life Span and Delays Rising old in Mice thru the Law of Nk2 Homeobox 1 within the DMH and LH. Cell Metabolism. 2013;18(3): 416–430. WOS: 000326267300014. pmid: 24011076 - 7.
Kim KW, Sohn JW, Kohno D, Xu Y, Williams Okay, Elmquist JK. SF-1 within the ventral medial hypothalamic nucleus: a key regulator of homeostasis. Mol Cell Endocrinol. 2011;336(1–2): 219–23. Epub 2010/11/30. pmid: 21111025; PubMed Central PMCID: PMC3057357. - 8.
Tang Y, Purkayastha S, Cai D. Hypothalamic microinflammation: a stylish foundation of metabolic syndrome and rising old. Trends Neurosci. 2015;38(1): 36–44. Epub 2014/12/03. pmid: 25458920; PubMed Central PMCID: PMC4282817. - 9.
Cai D, Khor S. “Hypothalamic Microinflammation” Paradigm in Rising old and Metabolic Ailments. Cell Metab. 2019;30(1): 19–35. Epub 2019/07/04. pmid: 31269425. - 10.
Kleinridders A, Schenten D, Konner AC, Belgardt BF, Mauer J, Okamura T, et al. MyD88 signaling within the CNS is required for vogue of fatty acid-induced leptin resistance and weight reduction program-induced obesity. Cell Metab. 2009;10(4): 249–59. Epub 2009/10/08. pmid: 19808018; PubMed Central PMCID: PMC3898351. - 11.
Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, et al. Saturated fatty acids develop an inflammatory response predominantly thru the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. J Neurosci. 2009;29(2): 359–70. Epub 2009/01/16. pmid: 19144836; PubMed Central PMCID: PMC6664935. - 12.
Kim MS, Yan J, Wu W, Zhang G, Zhang Y, Cai D. Quick linkage of innate immunological alerts to adaptive immunity by the brain-plump axis. Nat Immunol. 2015;16(5): 525–33. Epub 2015/04/08. pmid: 25848866; PubMed Central PMCID: PMC4564120. - 13.
Timper Okay, Denson JL, Steculorum SM, Heilinger C, Engstrom-Ruud L, Wunderlich CM, et al. IL-6 Improves Energy and Glucose Homeostasis in Obesity thru Enhanced Central IL-6 trans-Signaling. Cell Safe. 2017;19(2): 267–80. Epub 2017/04/14. pmid: 28402851. - 14.
Leng L, Zhuang Okay, Liu Z, Huang C, Gao Y, Chen G, et al. Menin Deficiency Outcomes in Depressive-esteem Behaviors in Mice by Modulating Astrocyte-Mediated Neuroinflammation. Neuron. 2018;100(3): 551–63 e7. Epub 2018/09/18. pmid: 30220511. - 15.
Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, et al. Leptin right away activates SF1 neurons within the VMH, and this streak by leptin is required for stylish physique-weight homeostasis. Neuron. 2006;49(2): 191–203. Epub 2006/01/21. pmid: 16423694. - 16.
Kim KW, Donato J Jr, Berglund ED, Choi YH, Kohno D, Elias CF, et al. FOXO1 within the ventromedial hypothalamus regulates energy balance. J Clin Invest. 2012;122(7): 2578–89. Epub 2012/06/02. pmid: 22653058; PubMed Central PMCID: PMC3386826. - 17.
Xu Y, Hill JW, Fukuda M, Gautron L, Sohn JW, Kim KW, et al. PI3K signaling within the ventromedial hypothalamic nucleus is required for stylish energy homeostasis. Cell Metab. 2010;12(1): 88–95. Epub 2010/07/14. pmid: 20620998; PubMed Central PMCID: PMC2919367. - 18.
Senn SS, Le Foll C, Whiting L, Tarasco E, Duffy S, Lutz TA, et al. Unsilencing of native LepRs in hypothalamic SF1 neurons does no longer rescue overweight phenotype in LepR-deficient mice. Am J Physiol Regul Integr Comp Physiol. 2019;317(3):R451–R60. Epub 2019/07/18. pmid: 31314542. - 19.
Scacheri PC, Crabtree JS, Kennedy AL, Swain GP, Ward JM, Marx SJ, et al. Homozygous loss of menin is successfully tolerated in liver, a tissue no longer affected in MEN1. Mamm Genome. 2004;15(11): 872–7. Epub 2005/01/28. pmid: 15672591. - 20.
Luo X, Ikeda Y, Lala DS, Baity LA, Meade JC, Parker KL. A cell-specific nuclear receptor plays mandatory roles in adrenal and gonadal vogue. Endocr Res. 1995;21(1–2): 517–24. Epub 1995/02/01. pmid: 7588417. - 21.
Luo X, Ikeda Y, Parker KL. The cell-specific nuclear receptor steroidogenic component 1 plays a couple of roles in reproductive aim. Philos Trans R Soc Lond B Biol Sci. 1995;350(1333): 279–83. Epub 1995/11/29. pmid: 8570692. - 22.
Solar XL, Weckwerth W. COVAIN: a toolbox for uni- and multivariate statistics, time-sequence and correlation community diagnosis and inverse estimation of the differential Jacobian from metabolomics covariance records. Metabolomics. 2012;8(1):S81–S93. WOS: 000311683000008. - 23.
Wilson JL, Nagele T, Linke M, Demel F, Fritsch SD, Mayr HK, et al. Inverse Knowledge-Pushed Modeling and Multiomics Prognosis Unearths Phgdh as a Metabolic Checkpoint of Macrophage Polarization and Proliferation. Cell Safe. 2020;30(5): 1542–52 e7. Epub 2020/02/06. pmid: 32023468; PubMed Central PMCID: PMC7003064. - 24.
Amelio I, Cutruzzola F, Antonov A, Agostini M, Melino G. Serine and glycine metabolism in most cancers. Trends Biochem Sci. 2014;39(4): 191–8. Epub 2014/03/25. pmid: 24657017; PubMed Central PMCID: PMC3989988. - 25.
Zhuang Okay, Huang C, Leng L, Zheng H, Gao Y, Chen G, et al. Neuron-Explicit Menin Deletion Outcomes in Synaptic Dysfunction and Cognitive Impairment by Modulating p35 Expression. Cell Safe. 2018;24(3): 701–12. Epub 2018/07/19. pmid: 30021166; PubMed Central PMCID: PMC6434950. - 26.
Otte DM, Barcena de Arellano ML, Bilkei-Gorzo A, Albayram O, Imbeault S, Jeung H, et al. Effects of Power D-Serine Elevation on Animal Devices of Depression and Scare-Associated Behavior. PLoS ONE. 2013;8(6):e67131. Epub 2013/06/28. pmid: 23805296; PubMed Central PMCID: PMC3689701. - 27.
Matkar S, Thiel A, Hua X. Menin: a scaffold protein that controls gene expression and cell signaling. Trends Biochem Sci. 2013;38(8): 394–402. Epub 2013/07/16. pmid: 23850066; PubMed Central PMCID: PMC3741089. - 28.
Katona BW, Hojnacki T, Glynn RA, Paulosky KE, Szigety KM, Cao Y, et al. Menin-mediated Repression of Glycolysis in Combination with Autophagy Protects Colon Most cancers Against Runt-molecule EGFR Inhibitors. Mol Most cancers Ther. 2020;19(11): 2319–29. Epub 2020/09/04. pmid: 32879052; PubMed Central PMCID: PMC7921201. - 29.
Svoboda LK, Teh SSK, Sud S, Kerk S, Zebolsky A, Treichel S, et al. Menin regulates the serine biosynthetic pathway in Ewing sarcoma. J Pathol. 2018;245(3): 324–36. Epub 2018/04/20. pmid: 29672864; PubMed Central PMCID: PMC6388767. - 30.
Liu S, Solar Y, Jiang M, Li Y, Tian Y, Xue W, et al. Glyceraldehyde-3-phosphate dehydrogenase promotes liver tumorigenesis by modulating phosphoglycerate dehydrogenase. Hepatology. 2017;66(2): 631–45. Epub 2017/04/08. pmid: 28387968. - 31.
Basu AC, Tsai GE, Ma CL, Ehmsen JT, Mustafa AK, Han L, et al. Focused disruption of serine racemase affects glutamatergic neurotransmission and habits. Mol Psychiatry. 2009;14(7): 719–27. Epub 2008/12/10. pmid: 19065142; PubMed Central PMCID: PMC2786989. - 32.
Fossat P, Turpin FR, Sacchi S, Dulong J, Shi T, Rivet JM, et al. Glial D-serine gates NMDA receptors at excitatory synapses in prefrontal cortex. Cereb Cortex. 2012;22(3): 595–606. Epub 2011/06/22. pmid: 21690263. - 33.
Le Bail M, Martineau M, Sacchi S, Yatsenko N, Radzishevsky I, Conrod S, et al. Identification of the NMDA receptor coagonist is synapse specific and developmentally regulated within the hippocampus. Proc Natl Acad Sci U S A. 2015;112(2):E204–13. Epub 2015/01/01. pmid: 25550512; PubMed Central PMCID: PMC4299231. - 34.
Mothet JP, Parent AT, Wolosker H, Brady RO Jr, Linden DJ, Ferris CD, et al. D-serine is an endogenous ligand for the glycine philosophize of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci U S A. 2000;97(9): 4926–31. Epub 2000/04/26. pmid: 10781100; PubMed Central PMCID: PMC18334. - 35.
Papouin T, Ladepeche L, Ruel J, Sacchi S, Labasque M, Hanini M, et al. Synaptic and extrasynaptic NMDA receptors are gated by varied endogenous coagonists. Cell. 2012;150(3): 633–46. Epub 2012/08/07. pmid: 22863013. - 36.
Wolosker H. Serine racemase and the serine shuttle between neurons and astrocytes. Biochim Biophys Acta. 2011;1814(11): 1558–1566. WOS: 000296952800018. pmid: 21224019 - 37.
Benneyworth MA, Li Y, Basu AC, Bolshakov VY, Coyle JT. Cell Selective Conditional Null Mutations of Serine Racemase Level to a Predominate Localization in Cortical Glutamatergic Neurons. Cell Mol Neurobiol. 2012;32(4): 613–624. WOS: 000303410000014. pmid: 22362148 - 38.
Balu DT, Coyle JT. Neuronal D-serine regulates dendritic structure within the somatosensory cortex. Neurosci Lett. 2012;517(2): 77–81. WOS: 000304846600003. pmid: 22521586 - 39.
Balu DT, Presti KT, Huang CCY, Muszynski Okay, Radzishevsky I, Wolosker H, et al. Serine Racemase and D-serine within the Amygdala Are Dynamically Enthusiastic in Concern Discovering out. Biol Psychiat. 2018;83(3): 273–283. WOS: 000418690300013. pmid: 29025687 - 40.
Xing B, Ma J, Jiang Z, Feng Z, Ling S, Szigety Okay, et al. GLP-1 signaling suppresses menin’s transcriptional block by phosphorylation in beta cells. J Cell Biol. 2019;218(3): 855–70. Epub 2019/02/23. pmid: 30792230; PubMed Central PMCID: PMC6400573. - 41.
Mensah-Osman E, Zavros Y, Service provider JL. Somatostatin stimulates menin gene expression by inhibiting protein kinase A. Am J Physiol Gastrointest Liver Physiol. 2008;295(4):G843–54. Epub 2008/08/30. pmid: 18755809; PubMed Central PMCID: PMC2575917. - 42.
Crabtree JS, Scacheri PC, Ward JM, McNally SR, Swain GP, Montagna C, et al. Of mice and MEN1: Insulinomas in a conditional mouse knockout. Mol Cell Biol. 2003;23(17): 6075–6085: pmid: 12917331; PubMed Central PMCID: PMC180910. - 43.
Miles AR, Hawrysh PJ, Hossein-Javaheri N, Buck LT. Taurine activates glycine and GABAA receptor currents in anoxia-tolerant painted turtle pyramidal neurons. J Exp Biol. 2018;221(Pt 21). Epub 2018/09/22. pmid: 30237241.