{"title":"Frontiers in radiopharmaceuticals for neuroendocrine tumors.","authors":"Ameya D Puranik, Indraja D Dev, Vikas Prasad","doi":"10.1111/jne.70006","DOIUrl":"https://doi.org/10.1111/jne.70006","url":null,"abstract":"<p><p>Neuroendocrine tumors encompass a wide range of tumors which originate from neural crest cells. These tumors were thought to be rare tumors, however, with the advent of advanced diagnostic techniques along with better understanding of the clinical presentation and histology of these tumors, the incidence of these tumors is exponentially rising. As the incidence and detection rate of NENs increased, the concept of 'heterogeneity' came into picture, which in turn led to dual-tracer imaging with addition of FDG PET/CT. Despite an imaging-based decision-making approach for NENs, there is still a significant subset of patients where the imaging-based biomarkers fall short in disease assessment, prognostication and improving outcomes. Alternate pathways as well as better peptide vectors for targeting the somatostatin receptor need to be studied. In this article, we address the existing as well as emerging trends in radiopharmaceuticals used for NENs, which are likely to impact not just the diagnostic algorithms in future, but also management strategies.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70006"},"PeriodicalIF":3.3,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Single-cell genomics meets systems neuroscience: Insights from mapping the brain circuitry of stress.","authors":"Naresh K Hanchate","doi":"10.1111/jne.70005","DOIUrl":"https://doi.org/10.1111/jne.70005","url":null,"abstract":"<p><p>Responses to external and internal dangers is essential for survival and homeostatic regulation. Hypothalamic corticotropin-releasing hormone neurons (CRHNs) play a pivotal role in regulating neuroendocrine responses to fear and stress. In recent years, the application of neurogenetic tools, such as fiber photometry, chemogenetics and optogenetics, have provided new insights into the dynamic neuronal responses of CRHNs during stressful events, offering new perspectives into their functional significance in mediating neurobehavioural responses to stress. Transsynaptic viral tracers have facilitated the comprehensive mapping of neuronal inputs to CRHNs. Furthermore, the development and application of innovative single-cell genomic tools combined with viral tracing have begun to pave the way for a deeper understanding of the transcriptional profiles of neural circuit components, enabling molecular-anatomical circuit mapping. Here, I will discuss how these systems neuroscience approaches and novel single-cell genomic methods are advancing the molecular and functional mapping of stress neurocircuits, their associated challenges and future directions.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70005"},"PeriodicalIF":3.3,"publicationDate":"2025-02-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143433032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hallbera Gudmundsdottir, Rondell P Graham, Patricia T Greipp, Elizabeth B Habermann, Ryan A Knudson, Carrie A Brandt, Patrick Starlinger, Cornelius A Thiels, Susanne G Warner, Rory L Smoot, Mark J Truty, Michael L Kendrick, David M Nagorney, Sean P Cleary, Thorvardur R Halfdanarson
{"title":"Alternative lengthening of telomeres and Ki-67 proliferation index provide complementary information on recurrence risk after resection of pancreatic neuroendocrine tumors.","authors":"Hallbera Gudmundsdottir, Rondell P Graham, Patricia T Greipp, Elizabeth B Habermann, Ryan A Knudson, Carrie A Brandt, Patrick Starlinger, Cornelius A Thiels, Susanne G Warner, Rory L Smoot, Mark J Truty, Michael L Kendrick, David M Nagorney, Sean P Cleary, Thorvardur R Halfdanarson","doi":"10.1111/jne.70003","DOIUrl":"https://doi.org/10.1111/jne.70003","url":null,"abstract":"<p><p>Given the heterogeneous clinical behavior of pancreatic neuroendocrine tumors (pNETs), improved prognostic markers are needed to guide management and post-resection surveillance. Patients who underwent resection of large (≥3 cm) sporadic well-differentiated pNETs from 2000 to 2019 were identified. The Ki-67 proliferation index was determined using immunohistochemistry, and alternative lengthening of telomeres (ALT) status was assessed using fluorescence in situ hybridization. Recurrence-free and overall survival were estimated using Kaplan-Meier analysis. Multivariable Cox regression analysis evaluated factors associated with recurrence-free survival. A total of 106 patients were identified. ALT was positive in 57 (54%) and negative in 49 (46%). Ki-67 was ≥3% in 74 (70%) and <3% in 32 (30%). Tumors with Ki-67 ≥3% were more likely to be ALT positive (61% vs. 38%, p = .046). Stratifying by ALT status and Ki-67 proliferation index, median recurrence-free survival was 4.6 years for patients with ALT-positive/Ki-67 ≥3% tumors, 3.1 years for patients with ALT-positive/Ki-67 <3% tumors, 12.4 years for patients with ALT-negative/Ki-67 ≥3% tumors, and 20.2 years for patients with ALT-negative/Ki-67 <3% tumors (p < .001). Initial recurrence was distant in 82% and locoregional in 18%. Across all groups, overall survival was similar (p = .19). In multivariable analysis, advanced age, ALT positivity, perineural invasion, and lymph node metastases were associated with increased recurrence risk (all p < .05). ALT and Ki-67 provide complementary information on post-resection recurrence risk, which can guide subsequent surveillance and management strategies. These data support the incorporation of ALT testing into routine clinical practice.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70003"},"PeriodicalIF":3.3,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143408755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
M Kidd, I A Drozdov, A Chirindel, G Nicolas, D Imagawa, A Gulati, T Tsuchikawa, V Prasad, A B Halim, J Strosberg
{"title":"NETest® 2.0-A decade of innovation in neuroendocrine tumor diagnostics.","authors":"M Kidd, I A Drozdov, A Chirindel, G Nicolas, D Imagawa, A Gulati, T Tsuchikawa, V Prasad, A B Halim, J Strosberg","doi":"10.1111/jne.70002","DOIUrl":"https://doi.org/10.1111/jne.70002","url":null,"abstract":"<p><p>Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are challenging to diagnose and manage. Because there is a critical need for a reliable biomarker, we previously developed the NETest, a liquid biopsy test that quantifies the expression of 51 neuroendocrine tumor (NET)-specific genes in blood using real-time PCR (NETest 1.0). In this study, we have leveraged our well-established laboratory approach (blood collection, RNA isolation, qPCR) with contemporary supervised machine learning methods and expanded training and testing sets to improve the discrimination and calibration of the NETest algorithm (NETest 2.0). qPCR measurements of RNA-stabilized blood-derived gene expression of 51 NET markers were used to train two supervised classifiers. The first classifier trained on 78 Controls and 162 NETs, distinguishing NETs from controls; the second, trained on 134 stable disease samples, 61 progressive disease samples, differentiated stable from progressive NET disease. In all cases, 80% of data was retained for model training, while remaining 20% were used for performance evaluation. The predictive performance of the AI system was assessed using sensitivity, specificity, and Area under Received Operating Characteristic curves (AUROC). The algorithm with the highest performance was retained for validation in two independent validation sets. Validation Cohort #I consisted of 277 patients and 186 healthy controls from the United States, Latin America, Europe, Africa and Asia, while Validation Cohort #II comprised 291 European patients from the Swiss NET Registry. A specificity cohort of 147 gastrointestinal, pancreatic and lung malignancies (non-NETs) was also evaluated. NETest 2.0 Algorithm #1 (Random Forest/gene expression normalized to ATG4B) achieved an AUROC of 0.91 for distinguishing NETs from controls (Validation Cohort #I), with a sensitivity of 95% and specificity of 81%. In Validation Cohort #II, 92% of NETs with image-positive disease were detected. The AUROC for differentiating NETs from other malignancies was 0.95; the sensitivity was 92% and specificity 90%. NETest 2.0 Algorithm #2 (Random Forest/gene expression normalized to ALG9) demonstrated an AUROC of 0.81 in Validation Cohort #I and 0.82 in Validation Cohort #II for differentiating stable from progressive disease, with specificities of 81% and 82%, respectively. Model performance was not affected by gender, ethnicity or age. Substantial improvements in performance for both algorithms were identified in head-to-head comparisons with NETest 1.0 (diagnostic: p = 1.73 × 10<sup>-9</sup>; prognostic: p = 1.02 × 10<sup>-10</sup>). NETest 2.0 exhibited improved diagnostic and prognostic capabilities over NETest 1.0. The assay also demonstrated improved sensitivity for differentiating NETs from other gastrointestinal, pancreatic and lung malignancies. The validation of this tool in geographically diverse cohorts highlights their potential for widespread clinical use.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70002"},"PeriodicalIF":3.3,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143408822","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Magnus Andreas Nordstrand, Dordi Lea, Jon Arne Søreide
{"title":"Incidence of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): An updated systematic review of population-based reports from 2010 to 2023.","authors":"Magnus Andreas Nordstrand, Dordi Lea, Jon Arne Søreide","doi":"10.1111/jne.70001","DOIUrl":"https://doi.org/10.1111/jne.70001","url":null,"abstract":"<p><p>There is a general perception that the incidence of neuroendocrine neoplasms (NENs) has been increasing. Nevertheless, reports of actual population-based studies are scarce, and pertinent data from some geographical regions still need to be available. In this systematic literature review of population-based studies, we aimed to evaluate the available data to provide updated figures on the incidence of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). Guided by the PRISMA 2020 statement reporting items for systematic reviews, this study conducted a systematic search using Ovid in the bibliographic databases Embase, Medline, and Web of Science Core Collection. Only incidence-reporting studies were included. In total, 847 articles were identified, and through a strict evaluation process using predefined inclusion and exclusion criteria, we found 19 papers that reported the general incidence of GEP-NENs from all sites. In addition, we considered another 15 papers that focused on the epidemiologic aspects of single-organ studies. While the incidence rates of GEP-NEN vary across similar countries, the general incidence of GEP-NEN has been increasing worldwide in recent decades. The incidence of GEP-NENs has increased worldwide over the last two decades, and reliable figures from new regions add to this pattern. Nevertheless, variations in the classification, grading, and reporting of GEP-NENs in various studies make direct comparisons difficult.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70001"},"PeriodicalIF":3.3,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143399327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Michael A Morse, Erika J Crosby, Daniel M Halperin, Hope E Uronis, S David Hsu, Herbert I Hurwitz, Christel Rushing, Emily K Bolch, Dana A Warren, Ashley N Moyer, Melissa E Lowe, Donna Niedzwiecki
{"title":"Phase Ib/II study of Pembrolizumab with Lanreotide depot for advanced, progressive Gastroenteropancreatic neuroendocrine tumors (PLANET).","authors":"Michael A Morse, Erika J Crosby, Daniel M Halperin, Hope E Uronis, S David Hsu, Herbert I Hurwitz, Christel Rushing, Emily K Bolch, Dana A Warren, Ashley N Moyer, Melissa E Lowe, Donna Niedzwiecki","doi":"10.1111/jne.13496","DOIUrl":"https://doi.org/10.1111/jne.13496","url":null,"abstract":"<p><p>While performing a study of immune checkpoint blockade with the anti-PD-1 antibody pembrolizumab combined with the somatostatin analogue (SSA) lanreotide in patients with low- and intermediate-grade gastroenteropancreatic neuroendocrine tumors (GEP-NETs), we studied whether there were any immune correlates of response to the anti-PD-1 therapy that could guide future attempts to integrate immunotherapy into the treatment of NETs. Patients with grade 1 and 2 GEP-NETs who had progressed on a prior SSA received lanreotide 90 mg subcutaneously and pembrolizumab 200 mg intravenously every 3 weeks until progression or intolerable toxicity. Objective response rate (ORR) at any time in the study, clinical benefit rate (CBR, defined as stable disease or better), progression-free survival (PFS), and overall survival (OS) were measured. Changes in T cell subsets in peripheral blood before and during therapy were analyzed by multiparameter mass cytometry (CyTOF). Archived tissue samples were analyzed for PD-L1 expression and TIL infiltration. Twenty-two (22) patients (GI/pancreatic 14/8, median Ki67 7% [IQR 4, 10%], median 1.5 prior systemic therapies [range 1-4]) were enrolled. Among the GI-NETs, there was one partial response, the CBR was 50%, the median PFS was 8.5 months, and the median OS was 32.7 months. No responses were seen in pancreatic NETs, which had 0% CBR, a PFS of 2.7 months, and an OS of 23.9 months. Of the 16 analyzable tumors, 6 had detectable PD-L1 expression and 15 had detectable TILs. Neither TILs nor PD-L1 expression correlated with ORR or CBR. However, clinical benefit (SD or better) was associated with peripheral blood on-treatment effector memory T cell activation and progressive disease was associated with baseline peripheral blood regulatory T cell (Treg) activation. We conclude that immune checkpoint blockade had low activity in unselected patients with grade 1 and 2 GEP-NETs. Further study of strategies to reduce Treg activation or enhance effector memory activation during immunotherapy is warranted.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e13496"},"PeriodicalIF":3.3,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143399330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jesse M Lacasse, Nafissa Ismail, Patrizia Porcu, Ami P Raval
{"title":"Animal models of hormonal contraceptives: Understanding drug-specific and user-specific variables.","authors":"Jesse M Lacasse, Nafissa Ismail, Patrizia Porcu, Ami P Raval","doi":"10.1111/jne.13500","DOIUrl":"https://doi.org/10.1111/jne.13500","url":null,"abstract":"<p><p>This manuscript reviews findings from the symposium \"Hormonal Contraceptives and the Brain: A Focus on Rodent Models,\" presented at the 2024 meeting for Steroids and the Nervous System in Turin, Italy. Hormonal contraceptives (HCs) are widely used by over 300 million women globally, yet their neurobiological and behavioral impacts have only recently gained extensive research attention. This review emphasizes the importance of animal models in studying these effects due to the practical and ethical limitations of human studies. By distinguishing drug-specific variables (e.g., dosage, chemical composition, routes of administration) from user-specific variables (lifestyle factors, genetic predispositions), researchers can better understand HC-related outcomes. Here, we emphasize the utility of animal models for uncovering putative mechanisms underlying the effects of HCs observed in human studies. Moreover, the authors reflect on the design of the animal models of HC used in their experiments, past and present. We also discuss current research related to user-specific variables which highlight the vulnerability of adolescents to the adverse effects of HCs, exposure to stressors, and the compounded risks of HC when combined with substances like cannabinoids and nicotine. This review underscores the value of animal models in advancing our understanding of the broader neurobiological and behavioral impacts of HCs in humans. These studies are crucial for developing personalized medicine approaches and optimizing contraceptive regimens to mitigate risks.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e13500"},"PeriodicalIF":3.3,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143391021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bilge Albayrak, Giorgi Batsikadze, Lara Jablonski, Ursula Felderhoff-Müser, Tina Hörbelt-Grünheidt, Anna Lena Friedel, Raphael Hirtz, Katharina Heuser-Spura, Monia V Dewan
{"title":"The cortisol awakening response in very preterm born adults compared to term born adults.","authors":"Bilge Albayrak, Giorgi Batsikadze, Lara Jablonski, Ursula Felderhoff-Müser, Tina Hörbelt-Grünheidt, Anna Lena Friedel, Raphael Hirtz, Katharina Heuser-Spura, Monia V Dewan","doi":"10.1111/jne.70000","DOIUrl":"https://doi.org/10.1111/jne.70000","url":null,"abstract":"<p><p>Very preterm infants are at higher risk of long-term neurodevelopmental and psychiatric impairments, including anxiety. Prematurity is also linked to altered programming of the hypothalamus-pituitary-adrenal (HPA) axis, associated with stress-related diseases. The cortisol awakening response (CAR), marked by a rapid cortisol increase after waking, is a biomarker of HPA reactivity and is linked to psychiatric disorders. This study aimed to evaluate for the first time the CAR in adults born very preterm and to explore its association with anxiety and stress. Twenty-five young adults born very preterm and 24 age- and sex-matched term-born controls collected saliva samples on two consecutive mornings at 0, 30, 45, and 60 min after awakening. Anxiety was measured using the State-Trait Anxiety Inventory, and stress was assessed with the Perceived Stress Scale. The CAR was analyzed using the sample at 0 min (S1), total cortisol output (AUCg), and actual CAR (AUCi). There were no significant differences in AUCi. The preterm group exhibited lower S1 levels and a reduced AUCg. Preterm-born participants reported higher trait anxiety and stress, though no consistent link with the CAR was identified. Findings suggest long-term neuroendocrine changes in adults born very preterm, warranting further research. Clinical Trial Registration: Duetsche Register Klinischer Studien (DRKS): 00020235.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e70000"},"PeriodicalIF":3.3,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143391022","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Conserved DNA methylation signatures in the prefrontal cortex of female newborn and juvenile guinea pigs following antenatal betamethasone exposure.","authors":"Bona Kim, Alice Kostaki, Stephen G Matthews","doi":"10.1111/jne.13499","DOIUrl":"https://doi.org/10.1111/jne.13499","url":null,"abstract":"<p><p>Antenatal corticosteroids (ACS) improve perinatal survival when there is a risk of preterm birth. Although evidence suggests an increased risk of developing neurobehavioural disorders in exposed offspring, the mechanisms involved remain largely unknown. Here, we investigated the DNA methylation patterns in the prefrontal cortex (PFC) of ACS-exposed guinea pig offspring. We hypothesized that differential methylation will be evident at both newborn and juvenile ages. In two separate cohorts, pregnant guinea pigs were administered a subcutaneous injection of saline or betamethasone (1 mg/kg) on gestational days 50/51 to mimic a single course of ACS. The gDNA was isolated from the PFC of term-born female offspring on postnatal day 1 (PND1, n = 7/group) and PND14 (n = 6-7/group) to identify differentially methylated CpG sites (DMCs) using reduced representative bisulphite sequencing. In the PND1 PFC, 1521 DMCs, annotating 144 genes were identified following ACS. Identified genes are involved in pathways regulating 'developmental cellular process'. In the PND14 PFC, 776 DMCs representing 46 genes were identified and enriched in 'synaptic signalling' pathways. Though no individual DMCs were identified at both PND1 and PND14, differential methylation was consistently observed at the binding sites of transcription factors PLAGL1, TFAP2C, ZNF263 and SP1 at both ages. We have established that ACS exposure leads to altered DNA methylation in the PFC of guinea pig offspring at both newborn and juvenile ages. Notably, a unique methylation signature was consistently observed at four key transcription factor binding sites at both post-natal time points. These changes may predispose the development of altered neurobehavioural phenotypes that have been described in ACS-exposed offspring.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e13499"},"PeriodicalIF":3.3,"publicationDate":"2025-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143382657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Henryk F Urbanski, Maria-Luisa Appleman, Kristopher M Fecteau, David W Erikson, Steven G Kohama, Riley MacKinnon, Alejandro Lomniczi
{"title":"Pituitary gonadotropin-releasing hormone II as a possible mediator of positive estrogen feedback.","authors":"Henryk F Urbanski, Maria-Luisa Appleman, Kristopher M Fecteau, David W Erikson, Steven G Kohama, Riley MacKinnon, Alejandro Lomniczi","doi":"10.1111/jne.13498","DOIUrl":"10.1111/jne.13498","url":null,"abstract":"<p><p>It has previously been shown that rhesus macaques express two forms of gonadotropin-releasing hormone (GNRH1 and GNRH2) in the hypothalamus and that both forms can stimulate the release of luteinizing hormone (LH) in vivo. However, while much has been published about the role of GNRH1 in reproduction, very little is known about the hypophysiotropic function of GNRH2. To shed light on this issue, we studied the expression pattern of these two genes in different parts of the monkey hypothalamus and pituitary gland under controlled conditions of circulating estrogen levels, using qPCR, liquid chromatography with tandem mass spectrometry and RNAscope. GNRH1/GNRH1 expression was found throughout the hypothalamus and was largely unaffected by circulating estradiol levels. In contrast, GNRH2/GNRH2 expression was found to be enhanced by long-term treatment with estradiol and during the late follicular phase of the menstrual cycle, especially in the arcuate nucleus and pituitary gland. Together these findings suggest that pituitary GNRH2/GNRH2 (but not GNRH1/GNRH1) is induced by positive feedback-like levels of estradiol. This novel finding raises the possibility that GNRH2 plays a major role in triggering the preovulatory LH surge in primates, not only at the level of the hypothalamus but also the pituitary gland.</p>","PeriodicalId":16535,"journal":{"name":"Journal of Neuroendocrinology","volume":" ","pages":"e13498"},"PeriodicalIF":3.3,"publicationDate":"2025-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}