Journal of the Endocrine Society
Lizneva, D;Ievleva, K;Gumerova, A;Shelly, E;Korkmaz, F;Muradova, V;Netto, J;Kuo, T;Sultana, F;Kumar, P;Kramskiy, N;Ryu, V;Padilla, A;Hutchison, S;Yuen, T;Zaidi, M;
| DOI: 10.1210/jendso/bvac150.058
Menopause is associated with the loss of LH ovulatory surges and enhanced visceral adiposity. Visceral fat depots increase from 5-8% at premenopause to 15-20% of total body fat at postmenopause. Here, we report that high-dose LH, hCG, or small molecule LH/CGR agonist ORG43553 injected twice-a-week into 14-weeks-old C57BL/6 male mice protects them from diet-induced obesity. Testosterone levels were elevated in mice treated with LH or hCG, but not with ORG43553. Notably, the anti-obesity action of LH/hCG is independent of testosterone, as blocking the androgen receptor using flutamide yielded similar results. Importantly, male Lhcgr knockout mice on a high-fat diet treated with LH failed to display a reduction in adiposity, confirming the in vivo specificity of action. Furthermore, our data phenocopied Lhcgr haploinsufficiency in mice. We confirmed the presence of Lhcgr in mouse genital and inguinal fat pads, adipose-derived stromal vascular cells, as well as in differentiated and undifferentiated 3T3-L1 murine adipocytes by qPCR, RNAscope in situ hybridization, and immunohistochemistry. Sanger sequencing showed that the extracellular domain of Lhcgr in genital fat depot was identical to the ovarian receptor. Similarly, we identified LHCGR in human subcutaneous and visceral fat depots. Binding of intraperitoneally injected AlexaFluor-488-labeled hCG was found not only in mouse ovary, but also in genital and subcutaneous fat pad, further confirming the presence of LHCGR in adipose tissue. This binding could be competitively displaced in 3T3-L1 cells using unlabeled hCG. LH, hCG and ORG43553 activated ERK1/2 in a dose-dependent manner in undifferentiated and differentiated 3T3-L1 cells, suggesting that the adipose LHCGR is fully functional. LH, hCG, and ORG43553 reduced adipogenic differentiation in 3T3-L1 cells, which is further confirmed by RNA sequencing. Moreover, we observed, that LH and hCG also alters several aspects of immune response in adipose tissue, including inflammatory response and adaptive immunity. In conclusion, we demonstrated that LH/CG receptors are present and fully functional in adipose tissue, and that high-dose intermittent activation of LHCGR in mouse fat depots protects mice from diet-induced obesity and modifies adipose tissue immune response. Presentation: Saturday, June 11, 2022 1:42 p.m. - 1:47 p.m., Monday, June 13, 2022 12:30 p.m. - 2:30 p.m.
Li, G;Xia, J;Zeng, W;Luo, W;Liu, L;Zeng, X;Cao, D;
| DOI: 10.3389/fimmu.2023.1206299
Located in the frontline against the largest population of microbiota, the intestinal mucosa of mammals has evolved to become an effective immune system. γδ T cells, a unique T cell subpopulation, are rare in circulation blood and lymphoid tissues, but rich in the intestinal mucosa, particularly in the epithelium. Via rapid production of cytokines and growth factors, intestinal γδ T cells are key contributors to epithelial homeostasis and immune surveillance of infection. Intriguingly, recent studies have revealed that the intestinal γδ T cells may play novel exciting functions ranging from epithelial plasticity and remodeling in response to carbohydrate diets to the recovery of ischemic stroke. In this review article, we update regulatory molecules newly defined in lymphopoiesis of the intestinal γδ T cells and their novel functions locally in the intestinal mucosa, such as epithelial remodeling, and distantly in pathological setting, e.g., ischemic brain injury repair, psychosocial stress responses, and fracture repair. The challenges and potential revenues in intestinal γδ T cell studies are discussed.
Zalachoras, I;Astori, S;Meijer, M;Grosse, J;Zanoletti, O;de Suduiraut, IG;Deussing, JM;Sandi, C;
PMID: 35319997 | DOI: 10.1126/sciadv.abj9019
Individuals frequently differ in their behavioral and cognitive responses to stress. However, whether motivation is differently affected by acute stress in different individuals remains to be established. By exploiting natural variation in trait anxiety in outbred Wistar rats, we show that acute stress facilitates effort-related motivation in low anxious animals, while dampening effort in high anxious ones. This model allowed us to address the mechanisms underlying acute stress-induced differences in motivated behavior. We show that CRHR1 expression levels in dopamine neurons of the ventral tegmental area (VTA)-a neuronal type implicated in the regulation of motivation-depend on animals' anxiety, and these differences in CRHR1 expression levels explain the divergent effects of stress on both effortful behavior and the functioning of mesolimbic DA neurons. These findings highlight CRHR1 in VTA DA neurons-whose levels vary with individuals' anxiety-as a switching mechanism determining whether acute stress facilitates or dampens motivation.
Examining Neuronal Subtype-Specific Genes Downregulated in Schizophrenia (SCZ) Using RNAscope in the Dorso-Lateral Prefrontal Cortex (DLPFC)
Kim, D;Jambhale, A;Kim, P;Kim, B;Lipska, B;Auluck, P;Marenco, S;
| DOI: 10.1016/j.biopsych.2021.02.630
Background Bulk tissue RNA-sequencing studies in human post-mortem brains have reported differentially expressed genes between patients with Schizophrenia (SCZ) and controls. Single-nucleus RNA-sequencing data indicate that genes downregulated in SCZ were particularly enriched in a sub-population of excitatory neurons (Ex21) indexed by SMYD1 gene. POSTN and NR4A2 are two genes highly specific to Ex21 and significantly downregulated in SCZ. We hypothesized that either the number of Ex21 cells or the expression of POSTN and NR4A2 within them would be reduced in SCZ vs. controls. Methods We conducted a RNAscope in situ hybridization study in 20 SCZ and 20 controls. Three sections of the DLPFC per individual were stained for SMYD1 and NR4A2, and another three for SMYD1 and POSTN, using the colorimetric version of RNAscope (240 sections). Each RNAscope slide was counterstained using hematoxylin/eosin. Additional sections were stained with NeuN to assess neuronal numbers. Results ANCOVA revealed that there were no significant differences between SCZ and controls in the proportion of Ex21 (SMYD1+) cells (p = 0.40 for POSTN dataset, 0.36 for NR4A2 dataset), or the expression of marker and target genes within SMYD1+ cells in the DLPFC (p = 0.86 for POSTN dataset, p = 0.15 for NR4A2 dataset). Conclusions These results contradict prior conclusions that both POSTN and NR4A2 are downregulated in SCZ, but the modest sample size and several methodological issues suggest caution in interpreting these results.
Wang, B;Jiang, B;Li, GW;Dong, F;Luo, Z;Cai, B;Wei, M;Huang, J;Wang, K;Feng, X;Tong, F;Wang, S;Wang, Q;Han, Q;Li, C;Zhang, X;Yang, L;Bao, L;
PMID: 36524339 | DOI: 10.15252/embr.202154313
Somatosensory neurons are highly heterogeneous with distinct types of neural cells responding to specific stimuli. However, the distribution and roles of cell-type-specific long intergenic noncoding RNAs (lincRNAs) in somatosensory neurons remain largely unexplored. Here, by utilizing droplet-based single-cell RNA-seq (scRNA-seq) and full-length Smart-seq2, we show that lincRNAs, but not coding mRNAs, are enriched in specific types of mouse somatosensory neurons. Profiling of lincRNAs from single neurons located in dorsal root ganglia (DRG) identifies 200 lincRNAs localized in specific types or subtypes of somatosensory neurons. Among them, the conserved cell-type-specific lincRNA CLAP associates with pruritus and is abundantly expressed in somatostatin (SST)-positive neurons. CLAP knockdown reduces histamine-induced Ca2+ influx in cultured SST-positive neurons and in vivo reduces histamine-induced scratching in mice. In vivo knockdown of CLAP also decreases the expression of neuron-type-specific and itch-related genes in somatosensory neurons, and this partially depends on the RNA binding protein MSI2. Our data reveal a cell-type-specific landscape of lincRNAs and a function for CLAP in somatosensory neurons in sensory transmission.
Kawai, K;Sakamoto, A;Mokry, M;Ghosh, S;Xu, W;
| DOI: 10.1161/circ.146.suppl_1.15249
Background: Studies using techniques that relied on expression of an X-linked gene suggested predominant clones of smooth muscle cells (SMC) may exist in human atherosclerosis. These studies were limited by spatial resolution and nature of plaque types studied. We investigated whether clones of SMCs exist in unstable human atheroma. Methods and Results: We used a 25 nucleotide deletion in the 3’ UTR of the BGN gene, highly expressed by SMC and prevalent in 30% of females, to study clonal proliferation. Three different types of plaques (erosion, rupture, and adaptive intimal thickening) were selected from females heterozygous for the deletion mutant. Hybridization of target RNA-specific BaseScope probes was conducted to visualize the distribution of mutants and images displayed as a bubble plots. Clonality index was calculated as the percentage of each probe in each ROI. A clonality index equal to or exceeding the three times the standard deviation above the mean of the clonality index of the media in all plaques was considered clonal. In comparing clonality between media and intima, the mean percent ROI with clonality was significantly higher in the intima than in the media (42.3±18.2 vs 18.3±9.6%, P=0.003) and this was consistent for both eroded (27.0±9.8 vs 9.0±3.8%, P=0.04) and ruptured plaques (41.3±10.7 vs 20.0±3.5%, P=0.03). The relationship of dominant clone in the intima and media shows significant concordance in the majority of plaques studied (R=0.72, P
Fang, YP;Qin, ZH;Zhang, Y;Ning, B;
PMID: 36216123 | DOI: 10.1016/j.expneurol.2022.114239
Microglia are widely distributed in the central nervous system (CNS), where they aid in the maintenance of neuronal function and perform key auxiliary roles in phagocytosis, neural repair, immunological control, and nutrition delivery. Microglia in the undamaged spinal cord is in a stable state and serve as immune monitors. In the event of spinal cord injury (SCI), severe changes in the microenvironment and glial scar formation lead to axonal regeneration failure. Microglia participates in a series of pathophysiological processes and behave both positive and negative consequences during this period. A deep understanding of the characteristics and functions of microglia can better identify therapeutic targets for SCI. Technological innovations such as single-cell RNA sequencing (Sc-RNAseq) have led to new advances in the study of microglia heterogeneity throughout the lifespan. Here,We review the updated studies searching for heterogeneity of microglia from the developmental and pathological state, survey the activity and function of microglia in SCI and explore the recent therapeutic strategies targeting microglia in the CNS injury.
Porcu, A;Nilsson, A;Booreddy, S;Barnes, SA;Welsh, DK;Dulcis, D;
PMID: 36054362 | DOI: 10.1126/sciadv.abn9867
Seasonal changes in day length (photoperiod) affect numerous physiological functions. The suprachiasmatic nucleus (SCN)-paraventricular nucleus (PVN) axis plays a key role in processing photoperiod-related information. Seasonal variations in SCN and PVN neurotransmitter expression have been observed in humans and animal models. However, the molecular mechanisms by which the SCN-PVN network responds to altered photoperiod is unknown. Here, we show in mice that neuromedin S (NMS) and vasoactive intestinal polypeptide (VIP) neurons in the SCN display photoperiod-induced neurotransmitter plasticity. In vivo recording of calcium dynamics revealed that NMS neurons alter PVN network activity in response to winter-like photoperiod. Chronic manipulation of NMS neurons is sufficient to induce neurotransmitter switching in PVN neurons and affects locomotor activity. Our findings reveal previously unidentified molecular adaptations of the SCN-PVN network in response to seasonality and the role for NMS neurons in adjusting hypothalamic function to day length via a coordinated multisynaptic neurotransmitter switching affecting behavior.
Science translational medicine
Liu, J;Trefry, JC;Babka, AM;Schellhase, CW;Coffin, KM;Williams, JA;Raymond, JLW;Facemire, PR;Chance, TB;Davis, NM;Scruggs, JL;Rossi, FD;Haddow, AD;Zelko, JM;Bixler, SL;Crozier, I;Iversen, PL;Pitt, ML;Kuhn, JH;Palacios, G;Zeng, X;
PMID: 35138912 | DOI: 10.1126/scitranslmed.abi5229
Effective therapeutics have been developed against acute Ebola virus disease (EVD) in both humans and experimentally infected nonhuman primates. However, the risk of viral persistence and associated disease recrudescence in survivors receiving these therapeutics remains unclear. In contrast to rhesus macaques that survived Ebola virus (EBOV) exposure in the absence of treatment, we discovered that EBOV, despite being cleared from all other organs, persisted in the brain ventricular system of rhesus macaque survivors that had received monoclonal antibody (mAb) treatment. In mAb-treated macaque survivors, EBOV persisted in macrophages infiltrating the brain ventricular system, including the choroid plexuses. This macrophage infiltration was accompanied by severe tissue damage, including ventriculitis, choroid plexitis, and meningoencephalitis. Specifically, choroid plexus endothelium-derived EBOV infection led to viral persistence in the macaque brain ventricular system. This resulted in apoptosis of ependymal cells, which constitute the blood-cerebrospinal fluid barrier of the choroid plexuses. Fatal brain-confined recrudescence of EBOV infection manifested as severe inflammation, local pathology, and widespread infection of the ventricular system and adjacent neuropil in some of the mAb-treated macaque survivors. This study highlights organ-specific EBOV persistence and fatal recrudescent disease in rhesus macaque survivors after therapeutic treatment and has implications for the long-term follow-up of human survivors of EVD.
Progress in retinal and eye research
Lewandowski, D;Sander, CL;Tworak, A;Gao, F;Xu, Q;Skowronska-Krawczyk, D;
PMID: 34971765 | DOI: 10.1016/j.preteyeres.2021.101037
The retinal pigment epithelium-photoreceptor interphase is renewed each day in a stunning display of cellular interdependence. While photoreceptors use photosensitive pigments to convert light into electrical signals, the RPE supports photoreceptors in their function by phagocytizing shed photoreceptor tips, regulating the blood retina barrier, and modulating inflammatory responses, as well as regenerating the 11-cis-retinal chromophore via the classical visual cycle. These processes involve multiple protein complexes, tightly regulated ligand-receptors interactions, and a plethora of lipids and protein-lipids interactions. The role of lipids in maintaining a healthy interplay between the RPE and photoreceptors has not been fully delineated. In recent years, novel technologies have resulted in major advancements in understanding several facets of this interplay, including the involvement of lipids in phagocytosis and phagolysosome function, nutrient recycling, and the metabolic dependence between the two cell types. In this review, we aim to integrate the complex role of lipids in photoreceptor and RPE function, emphasizing the dynamic exchange between the cells as well as discuss how these processes are affected in aging and retinal diseases.
Seeker, LA;Williams, A;
PMID: 34860266 | DOI: 10.1007/s00401-021-02390-4
It is the centenary of the discovery of oligodendrocytes and we are increasingly aware of their importance in the functioning of the brain in development, adult learning, normal ageing and in disease across the life course, even in those diseases classically thought of as neuronal. This has sparked more interest in oligodendroglia for potential therapeutics for many neurodegenerative/neurodevelopmental diseases due to their more tractable nature as a renewable cell in the central nervous system. However, oligodendroglia are not all the same. Even from the first description, differences in morphology were described between the cells. With advancing techniques to describe these differences in human tissue, the complexity of oligodendroglia is being discovered, indicating apparent functional differences which may be of critical importance in determining vulnerability and response to disease, and targeting of potential therapeutics. It is timely to review the progress we have made in discovering and understanding oligodendroglial heterogeneity in health and neuropathology.
The role of endoplasmic reticulum stress in astrocytes
Sims, SG;Cisney, RN;Lipscomb, MM;Meares, GP;
PMID: 34462963 | DOI: 10.1002/glia.24082
Astrocytes are glial cells that support neurological function in the central nervous system (CNS), in part, by providing structural support for neuronal synapses and blood vessels, participating in electrical and chemical transmission, and providing trophic support via soluble factors. Dysregulation of astrocyte function contributes to neurological decline in CNS diseases. Neurological diseases are highly heterogeneous but share common features of cellular stress including the accumulation of misfolded proteins. Endoplasmic reticulum (ER) stress has been reported in nearly all neurological and neurodegenerative diseases. ER stress occurs when there is an accumulation of misfolded proteins in the ER lumen and the protein folding demand of the ER is overwhelmed. ER stress initiates the unfolded protein response (UPR) to restore homeostasis by abating protein translation and, if the cell is irreparably damaged, initiating apoptosis. Although protein aggregation and misfolding in neurological disease has been well described, cell-specific contributions of ER stress and the UPR in physiological and disease states are poorly understood. Recent work has revealed a role for active UPR signaling that may drive astrocytes toward a maladaptive phenotype in various model systems. In response to ER stress, astrocytes produce inflammatory mediators, have reduced trophic support, and can transmit ER stress to other cells. This review will discuss the current known contributions and consequences of activated UPR signaling in astrocytes.