Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.644
Filter
1.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38952926

ABSTRACT

Background: Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. Methods: Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. Results: OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. Conclusions: SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.


Subject(s)
Disease Models, Animal , Extinction, Psychological , Fear , Mice, Knockout , Neurons , Oxytocin , Prader-Willi Syndrome , Somatostatin , Vasopressins , Animals , Oxytocin/pharmacology , Somatostatin/pharmacology , Somatostatin/metabolism , Fear/drug effects , Fear/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Neurons/drug effects , Neurons/metabolism , Mice , Prader-Willi Syndrome/physiopathology , Prader-Willi Syndrome/drug therapy , Vasopressins/metabolism , Aggression/drug effects , Aggression/physiology , Male , Social Behavior , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Optogenetics , Mice, Inbred C57BL , Intracellular Signaling Peptides and Proteins , Intrinsically Disordered Proteins
2.
Int J Mol Sci ; 25(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-39000000

ABSTRACT

Somatostatin (SS) plays crucial regulatory roles in animal growth and reproduction by affecting the synthesis and secretion of growth hormone (GH). However, the mechanism by which SS regulates growth and development in goats is still unclear. In order to investigate the regulatory networks of the hypothalamus and pituitary in goats affected by SS DNA vaccines, in this study, we used a previously established oral attenuated Salmonella typhimurium SS DNA vaccine, X9241 (ptCS/2SS-asd), to treat wethers. We analyzed the protein changes in hypothalamic and pituitary tissues using a TMT-based proteomics approach. Additionally, we examined the metabolic profiles of the serum of control and immunized wethers through untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS). Key signaling pathways were identified based on differentially expressed metabolites (DEMs) and differentially expressed proteins (DEPs). Furthermore, the effect of critical DEPs on signaling pathways was confirmed through Western blotting (WB) experiments, which elucidated the mechanism of active SS immunization in wethers. A proteomics analysis revealed that the expression of 58 proteins in the hypothalamus and 124 in the pituitary gland was significantly altered following SS vaccine treatment (fold change > 1.2 or < 0.83, p < 0.05). In the hypothalamus, many DEPs were associated with gene ontology (GO) terms related to neuronal signaling. In contrast, most DEPs were associated with metabolic pathways. In the pituitary gland, the DEPs were largely related to immune and nutrient metabolism functions, with significant enrichment in KEGG pathways, particularly those involving the metabolic pathway, sphingolipid signaling, and the cGMP-PKG signaling pathway. A metabolomic analysis further showed that active SS immunization in wethers led to significant alterations in seven serum metabolites. Notably, the sphingolipid signaling pathway, secondary bile acid synthesis, sphingolipid metabolism, and lysine synthesis were significantly disrupted. SS vaccines induced marked changes in hypothalamic-pituitary proteins in wethers, facilitating alterations in their growth processes. This study not only provides insights into the mechanism of the SS gene in regulating GH secretion in wethers but also establishes a basis for hormone immunoregulation technology to enhance livestock production performance.


Subject(s)
Goats , Hypothalamus , Pituitary Gland , Proteomics , Somatostatin , Vaccines, DNA , Animals , Somatostatin/metabolism , Proteomics/methods , Hypothalamus/metabolism , Vaccines, DNA/immunology , Pituitary Gland/metabolism , Metabolomics/methods , Signal Transduction , Metabolome
3.
Front Endocrinol (Lausanne) ; 15: 1302672, 2024.
Article in English | MEDLINE | ID: mdl-38974572

ABSTRACT

Somatostatin (SST) plays diverse physiological roles in vertebrates, particularly in regulating growth hormone secretion from the pituitary. While the function of SST as a neuromodulator has been studied extensively, its role in fish and mammalian reproduction remains poorly understood. To address this gap, we investigated the involvement of the somatostatin system in the regulation of growth and reproductive hormones in tilapia. RNA sequencing of mature tilapia brain tissue revealed the presence of three SST peptides: SST6, SST3, and low levels of SST1. Four different isoforms of the somatostatin receptor (SSTR) subfamily were also identified in the tilapia genome. Phylogenetic and synteny analysis identified tiSSTR2-like as the root of the tree, forming two mega clades, with SSTR1 and SSTR4 in one and SSTR2a, SSTR3a, and SSTR5b in the other. Interestingly, the tiSSTR-5 isoforms 5x1, 5x2, and 5x3 were encoded in the sstr3b gene and were an artifact of misperception in the nomenclature in the database. RNA-seq of separated pituitary cell populations showed that SSTRs were expressed in gonadotrophs, with sstr3a enriched in luteinizing hormone (LH) cells and sstr3b significantly enriched in follicle-stimulating hormone (FSH) cells. Notably, cyclosomatostatin, an SSTR antagonist, induced cAMP activity in all SSTRs, with SSTR3a displaying the highest response, whereas octreotide, an SSTR agonist, showed a binding profile like that observed in human receptors. Binding site analysis of tiSSTRs from tilapia pituitary cells revealed the presence of canonical binding sites characteristic of peptide-binding class A G-protein-coupled receptors. Based on these findings, we explored the effect of somatostatin on gonadotropin release from the pituitary in vivo. Whereas cyclosomatostatin increased LH and FSH plasma levels at 2 h post-injection, octreotide decreased FSH levels after 2 h, but the LH levels remained unaffected. Overall, our findings provide important insights into the somatostatin system and its mechanisms of action, indicating a potential role in regulating growth and reproductive hormones. Further studies of the complex interplay between SST, its receptors, and reproductive hormones may advance reproductive control and management in cultured populations.


Subject(s)
Phylogeny , Receptors, Somatostatin , Reproduction , Somatostatin , Tilapia , Animals , Tilapia/metabolism , Tilapia/growth & development , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/genetics , Somatostatin/metabolism , Reproduction/physiology , Pituitary Gland/metabolism , Humans , Female , Male
4.
Front Endocrinol (Lausanne) ; 15: 1385079, 2024.
Article in English | MEDLINE | ID: mdl-38948517

ABSTRACT

Background: 177Lu-oxodotreotide peptide receptor therapy (LuPRRT) is an efficient treatment for midgut neuroendocrine tumors (NETs) of variable radiological response. Several clinical, biological, and imaging parameters may be used to establish a relative disease prognosis but none is able to predict early efficacy or toxicities. We investigated expression levels for mRNA and miRNA involved in radiosensitivity and tumor progression searching for correlations related to patient outcome during LuPRRT therapy. Methods: Thirty-five patients received LuPRRT for G1/G2 midgut NETs between May 2019 and September 2021. Peripheral blood samples were collected prior to irradiation, before and 48 h after the second and the fourth LuPRRT, and at 6-month follow-up. Multiple regression analyses and Pearson correlations were performed to identify the miRNA/mRNA signature that will best predict response to LuPRRT. Results: Focusing on four mRNAs and three miRNAs, we identified a miRNA/mRNA signature enabling the early identification of responders to LuPRRT with significant reduced miRNA/mRNA expression after the first LuPRRT administration for patients with progressive disease at 1 year (p < 0.001). The relevance of this signature was reinforced by studying its evolution up to 6 months post-LuPRRT. Moreover, nadir absolute lymphocyte count within the first 2 months after the first LuPRRT administration was significantly related to low miRNA/mRNA expression level (p < 0.05) for patients with progressive disease. Conclusion: We present a pilot study exploring a miRNA/mRNA signature that correlates with early hematologic toxicity and therapeutic response 12 months following LuPRRT. This signature will be tested prospectively in a larger series of patients.


Subject(s)
Intestinal Neoplasms , MicroRNAs , Neuroendocrine Tumors , RNA, Messenger , Humans , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/pathology , Male , Female , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Intestinal Neoplasms/blood , Intestinal Neoplasms/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/drug therapy , RNA, Messenger/genetics , RNA, Messenger/blood , Aged , Follow-Up Studies , Adult , Prognosis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Receptors, Peptide/genetics , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/administration & dosage , Lutetium , Radioisotopes
5.
Dtsch Med Wochenschr ; 149(15): 871-878, 2024 Aug.
Article in German | MEDLINE | ID: mdl-39013407

ABSTRACT

Despite, or perhaps because of the rarity of neuroendocrine neoplasms, the diagnosis and treatment of these malignancies is of particular importance. Nuclear medicine can make an important contribution to this challenge. It offers the most sensitive and specific imaging of these tumor entities and can be helpful in treatment due to the radiotherapeutic drugs that have recently been approved. This theragnostic (fusion of therapeutic and diagnostic) concept is based on the frequent overexpression of somatostatin receptors on neuroendocrine tumor cells.Using diagnostic and therapeutic pharmaceuticals based on analogues from somatostatin, most applications from the nuclear medicine are successful, an additional therapeutic method is SIRT, also known as TARE, in which the hypervascularization of NEN-metastases is used as a therapeutic target.


Subject(s)
Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/therapy , Nuclear Medicine/methods , Radiopharmaceuticals/therapeutic use , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Receptors, Somatostatin
6.
Int J Nanomedicine ; 19: 4977-4994, 2024.
Article in English | MEDLINE | ID: mdl-38828204

ABSTRACT

Purpose: Exosomes are membrane vesicles secreted by various cells and play a crucial role in intercellular communication. They can be excellent delivery vehicles for oligonucleotide drugs, such as microRNAs, due to their high biocompatibility. MicroRNAs have been shown to be more stable when incorporated into exosomes; however, the lack of targeting and immune evasion is still the obstacle to the use of these microRNA-containing nanocarriers in clinical settings. Our goal was to produce functional exosomes loaded with target ligands, immune evasion ligand, and oligonucleotide drug through genetic engineering in order to achieve more precise medical effects. Methods: To address the problem, we designed engineered exosomes with exogenous cholecystokinin (CCK) or somatostatin (SST) as the targeting ligand to direct the exosomes to the brain, as well as transduced CD47 proteins to reduce the elimination or phagocytosis of the targeted exosomes. MicroRNA-29b-2 was the tested oligonucleotide drug for delivery because our previous research showed that this type of microRNA was capable of reducing presenilin 1 (PSEN1) gene expression and decreasing the ß-amyloid accumulation for Alzheimer's disease (AD) in vitro and in vivo. Results: The engineered exosomes, containing miR29b-2 and expressing SST and CD47, were produced by gene-modified dendritic cells and used in the subsequent experiments. In comparison with CD47-CCK exosomes, CD47-SST exosomes showed a more significant increase in delivery efficiency. In addition, CD47-SST exosomes led to a higher delivery level of exosomes to the brains of nude mice when administered intravenously. Moreover, it was found that the miR29b-2-loaded CD47-SST exosomes could effectively reduce PSEN1 in translational levels, which resulted in an inhibition of beta-amyloid oligomers production both in the cell model and in the 3xTg-AD animal model. Conclusion: Our results demonstrated the feasibility of the designed engineered exosomes. The application of this exosomal nanocarrier platform can be extended to the delivery of other oligonucleotide drugs to specific tissues for the treatment of diseases while evading the immune system.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , CD47 Antigen , Exosomes , MicroRNAs , Presenilin-1 , Receptors, Somatostatin , Animals , Exosomes/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , MicroRNAs/genetics , MicroRNAs/administration & dosage , Presenilin-1/genetics , Brain/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Amyloid beta-Peptides/metabolism , Mice , CD47 Antigen/genetics , CD47 Antigen/metabolism , Somatostatin , Humans , Disease Models, Animal
7.
Life Sci ; 351: 122854, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38901688

ABSTRACT

AIMS: To evaluate the cell proliferation and death, and structural morphology of the pancreatic islet cells of the rats with hyperglycemia in the first month of life and compare to those of the control rats. MAIN METHODS: Female Sprague-Dawley newborn rats received Streptozotocin (a beta-cytotoxic drug) at birth for diabetes induction. Control and hyperglycemic animals were euthanized on different days of life: 5, 10, 15, and 30. The pancreas was collected and processed for immunohistochemical analysis of cleaved Caspase-3 (cell death), Ki-67 (cell proliferation), PDX-1 (transcription factor responsible for insulin synthesis), and endocrine hormones (insulin, glucagon, and somatostatin). KEY FINDINGS: Control females showed a higher percentage (%) of Ki-67-positive(+) cells on D10 and D15, a higher % of insulin+ and somatostatin+ cells on D15 and D30, a lower % of PDX-1+ cells on D10, and a higher % of glucagon+ cells on D10 and D30. Hyperglycemic females showed a lower % of Ki-67+ cells on D15, a higher % of cleaved Caspase-3+ cells on D15, and insulin+ cells on D15 and D30. In the comparison among the experimental groups, the hyperglycemic females showed an increased % of cleaved Caspase-3+ and Ki-67+ cells and a lower % of PDX-1+ cells. SIGNIFICANCE: This study enabled a better understanding of the abnormal pancreas development regarding cellular proliferation, apoptosis, and hormonal synthesis in the neonatal period. Thus, the pancreatic islets of hyperglycemic rats do not reestablish the normal endocrine cell population, and cellular apoptosis overcame the proliferative activity of these cells.


Subject(s)
Animals, Newborn , Cell Proliferation , Hyperglycemia , Islets of Langerhans , Rats, Sprague-Dawley , Animals , Female , Hyperglycemia/metabolism , Hyperglycemia/pathology , Rats , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/metabolism , Cell Death , Glucagon/metabolism , Insulin/metabolism , Ki-67 Antigen/metabolism , Caspase 3/metabolism , Somatostatin/metabolism , Apoptosis , Trans-Activators , Homeodomain Proteins
8.
Org Lett ; 26(26): 5447-5452, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38896796

ABSTRACT

Interest in electrocatalytic bioconjugation reactions has surged, particularly for modifying tryptophan and tyrosine residues in proteins. We used a cost-effective graphite felt electrode and low-current methodology to achieve selective bioconjugation of tryptophan with thiophenols, yielding up to 92%. This method exclusively labeled tryptophan residues and incorporated fluorinated tryptophan for NMR analysis. Eight polypeptides, including lanreotide and leuprorelin, were effectively coupled, demonstrating the method's versatility and potential for novel diagnostic and therapeutic agents.


Subject(s)
Peptides , Tryptophan , Tryptophan/chemistry , Peptides/chemistry , Electrochemical Techniques , Molecular Structure , Somatostatin/chemistry , Somatostatin/analogs & derivatives , Peptides, Cyclic/chemistry , Electrodes
9.
Nat Commun ; 15(1): 5421, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926335

ABSTRACT

During brain development, neural circuits undergo major activity-dependent restructuring. Circuit wiring mainly occurs through synaptic strengthening following the Hebbian "fire together, wire together" precept. However, select connections, essential for circuit development, are transient. They are effectively connected early in development, but strongly diminish during maturation. The mechanisms by which transient connectivity recedes are unknown. To investigate this process, we characterize transient thalamocortical inputs, which depress onto somatostatin inhibitory interneurons during development, by employing optogenetics, chemogenetics, transcriptomics and CRISPR-based strategies in mice. We demonstrate that in contrast to typical activity-dependent mechanisms, transient thalamocortical connectivity onto somatostatin interneurons is non-canonical and involves metabotropic signaling. Specifically, metabotropic-mediated transcription, of guidance molecules in particular, supports the elimination of this connectivity. Remarkably, we found that this process impacts the development of normal exploratory behaviors of adult mice.


Subject(s)
Interneurons , Somatostatin , Thalamus , Animals , Interneurons/metabolism , Somatostatin/metabolism , Somatostatin/genetics , Mice , Thalamus/metabolism , Optogenetics , Signal Transduction , Male , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Female , Mice, Inbred C57BL , Mice, Transgenic
10.
Proc Natl Acad Sci U S A ; 121(26): e2321710121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38885377

ABSTRACT

Somatostatin receptor 5 (SSTR5) is an important G protein-coupled receptor and drug target for neuroendocrine tumors and pituitary disorders. This study presents two high-resolution cryogenicelectron microscope structures of the SSTR5-Gi complexes bound to the cyclic neuropeptide agonists, cortistatin-17 (CST17) and octreotide, with resolutions of 2.7 Å and 2.9 Å, respectively. The structures reveal that binding of these peptides causes rearrangement of a "hydrophobic lock", consisting of residues from transmembrane helices TM3 and TM6. This rearrangement triggers outward movement of TM6, enabling Gαi protein engagement and receptor activation. In addition to hydrophobic interactions, CST17 forms conserved polar contacts similar to somatostatin-14 binding to SSTR2, while further structural and functional analysis shows that extracellular loops differently recognize CST17 and octreotide. These insights elucidate agonist selectivity and activation mechanisms of SSTR5, providing valuable guidance for structure-based drug development targeting this therapeutically relevant receptor.


Subject(s)
Octreotide , Receptors, Somatostatin , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry , Humans , Octreotide/chemistry , Octreotide/pharmacology , Octreotide/metabolism , Neuropeptides/metabolism , Neuropeptides/chemistry , Cryoelectron Microscopy , Protein Binding , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/metabolism , Somatostatin/metabolism , Somatostatin/chemistry , Somatostatin/analogs & derivatives , Models, Molecular , HEK293 Cells
11.
Protein Expr Purif ; 222: 106537, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38944221

ABSTRACT

Peptides are used for diagnostics, therapeutics, and as antimicrobial agents. Most peptides are produced by chemical synthesis, but recombinant production has recently become an attractive alternative due to the advantages of high titers, less toxic waste and correct folding of tertiary structure. Somatostatin-28 is a peptide hormone that regulates the endocrine system, cell proliferation and inhibits the release of numerous secondary hormones in human body. It is composed of 28 amino acids and has one disulfide bond, which makes it to an optimal model peptide for a whole downstream purification process. We produced the peptide in the periplasm of E. coli using the CASPON™ technology, an affinity fusion technology system that enables high soluble expression of recombinant proteins and cleaves the fusion tag with a circularly permuted human caspase-2. Furthermore, purification of the products is straight forward using an established platform process. Two different case studies for downstream purification are presented, starting with either hydrochloric acid or polyethyleneimine as an extraction aid. After release of affinity-tagged somatostatin-28 out of E. coli's periplasm, several purification steps were performed, delivering a pure peptide solution after the final polishing step. The process was monitored by reversed-phase high-performance liquid chromatography as well as mass spectrometry to determine the yield and correct disulfide bond formation. Monitoring of impurities like host cell proteins, DNA and endotoxins after each downstream unit confirmed effective removal for both purification pathways.


Subject(s)
Escherichia coli , Hydrochloric Acid , Polyethyleneimine , Somatostatin , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Somatostatin/chemistry , Somatostatin/genetics , Somatostatin/isolation & purification , Hydrochloric Acid/chemistry , Polyethyleneimine/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/biosynthesis
12.
Expert Opin Drug Saf ; 23(8): 949-957, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38847075

ABSTRACT

INTRODUCTION: Neuroendocrine neoplasms (NENs) are a rare group of tumors originating from neuroendocrine cells in various organs. They include neuroendocrine tumors (NETs) and neuroendocrine carcinomas (NECs), which differ in biological behavior and prognosis. NETs are usually well-differentiated and slow-growing, while NECs are poorly differentiated and more aggressive. Management of NETs often involves somatostatin analogs like octreotide and lanreotide to control tumor growth and alleviate symptoms, especially in well-differentiated NETs. Lanreotide is used to control tumor growth, and both lanreotide and octreotide alleviate symptoms. Treatment approaches may vary depending on the specific type and grade of the neuroendocrine neoplasm. AREAS COVERED: This review provides an update on the safety of lanreotide autogel in treating patients with NETs, through a comprehensive review of clinical trials, post-marketing surveillance, real-world evidence, and its safety profile. Specific adverse events, side effects, and potential risks associated with lanreotide autogel are discussed, along with risk mitigation strategies and recommendations for patient monitoring. EXPERT OPINION: The findings highlight the overall safety of lanreotide autogel in managing NETs, focusing on its efficacy in controlling hormone secretion, tumor progression, and symptom management. New safety concerns and precautions are also addressed to help healthcare providers make informed decisions when prescribing lanreotide autogel.


Subject(s)
Antineoplastic Agents , Neuroendocrine Tumors , Peptides, Cyclic , Somatostatin , Humans , Somatostatin/analogs & derivatives , Somatostatin/administration & dosage , Somatostatin/adverse effects , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/pathology , Gels , Disease Progression
14.
Sci Rep ; 14(1): 13525, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866945

ABSTRACT

The traditional nomenclature of enteroendocrine cells (EECs), established in 1977, applied the "one cell - one hormone" dogma, which distinguishes subpopulations based on the secretion of a specific hormone. These hormone-specific subpopulations included S cells for secretin (SCT), K cells for glucose-dependent insulinotropic polypeptide (GIP), N cells producing neurotensin (NTS), I cells producing cholecystokinin (CCK), D cells producing somatostatin (SST), and others. In the past 15 years, reinvestigations into murine and human organoid-derived EECs, however, strongly questioned this dogma and established that certain EECs coexpress multiple hormones. Using the Gut Cell Atlas, the largest available single-cell transcriptome dataset of human intestinal cells, this study consolidates that the original dogma is outdated not only for murine and human organoid-derived EECs, but also for primary human EECs, showing that the expression of certain hormones is not restricted to their designated cell type. Moreover, specific analyses into SCT-expressing cells reject the presence of any cell population that exhibits significantly elevated secretin expression compared to other cell populations, previously referred to as S cells. Instead, this investigation indicates that secretin production is realized jointly by other enteroendocrine subpopulations, validating corresponding observations in murine EECs also for human EECs. Furthermore, our findings corroborate that SCT expression peaks in mature EECs, in contrast, progenitor EECs exhibit markedly lower expression levels, supporting the hypothesis that SCT expression is a hallmark of EEC maturation.


Subject(s)
Enteroendocrine Cells , Gene Expression Profiling , Secretin , Single-Cell Analysis , Humans , Enteroendocrine Cells/metabolism , Secretin/metabolism , Secretin/genetics , Single-Cell Analysis/methods , Mice , Animals , Transcriptome , Cell Differentiation , Organoids/metabolism , Organoids/cytology , Cholecystokinin/metabolism , Cholecystokinin/genetics , Somatostatin/metabolism , Somatostatin/genetics , Single-Cell Gene Expression Analysis
15.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791582

ABSTRACT

A novel nanotechnology-based drug delivery system (DDS) targeted at pancreatic cancer cells was developed, characterized, and tested. The system consisted of liposomes as carriers, an anticancer drug (paclitaxel) as a chemotherapeutic agent, and a modified synthetic somatostatin analog, 5-pentacarbonyl-octreotide, a ligand for somatostatin receptor 2 (SSTR2), as a targeting moiety for pancreatic cancer. The cellular internalization, cytotoxicity, and antitumor activity of the DDS were tested in vitro using human pancreatic ductal adenocarcinoma (PDAC) cells with different expressions of the targeted SSTR2 receptors, and in vivo on immunodeficient mice bearing human PDAC xenografts. The targeted drug delivery system containing paclitaxel exhibited significantly enhanced cytotoxicity compared to non-targeted DDS, and this efficacy was directly related to the levels of SSTR2 expression. It was found that octreotide-targeted DDS proved exceptionally effective in suppressing the growth of PDAC tumors. This study underscores the potential of octreotide-targeted liposomal delivery systems to enhance the therapeutic outcomes for PDAC compared with non-targeted liposomal DDS and Paclitaxel-Cremophor® EL, suggesting a promising avenue for future cancer therapy innovations.


Subject(s)
Drug Delivery Systems , Liposomes , Octreotide , Paclitaxel , Pancreatic Neoplasms , Receptors, Somatostatin , Xenograft Model Antitumor Assays , Animals , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Somatostatin/metabolism , Mice , Cell Line, Tumor , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Liposomes/chemistry , Drug Delivery Systems/methods , Octreotide/administration & dosage , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Nanotechnology/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology
16.
Expert Rev Gastroenterol Hepatol ; 18(4-5): 203-215, 2024.
Article in English | MEDLINE | ID: mdl-38725175

ABSTRACT

OBJECTIVE: To determine the effectiveness of the different pharmacological agents in preventing post-ERCP acute pancreatitis. METHODS: We included clinical trials of pharmacological interventions for prophylaxis of acute post-ERCP pancreatitis. The event evaluated was acute pancreatitis. We conducted a search strategy in MEDLINE (OVID), EMBASE, and Cochrane Central Register of Controlled Trials from inception to nowadays. We reported the information in terms of relative risks (RR) with a 95% confidence interval. We assessed the heterogeneity using the I2 test. RESULTS: We included 84 studies for analysis (30,463 patients). The mean age was 59.3 years (SD ± 7.01). Heterogeneity between studies was low (I2 = 34.4%) with no inconsistencies (p = 0.2567). Post ERCP pancreatitis was less in prophylaxis with NSAIDs (RR 0.65 95% CI [0.52 to 0.80]), aggressive hydration with Lactate Ringer (RR 0.32 95% CI [0.12-0.86]), NSAIDs + isosorbide dinitrate (RR 0.28 95% CI [0.11-0.71]) and somatostatin and analogues (RR 0.54 [0.43 to 0.68]) compared with placebo. CONCLUSIONS: NSAIDs, the Combination of NSAIDs + isosorbide dinitrate, somatostatin and analogues, and aggressive hydration with lactate ringer are pharmacological strategies that can prevent post-ERCP pancreatitis when compared to placebo. More clinical trials are required to determine the effectiveness of these drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Cholangiopancreatography, Endoscopic Retrograde , Pancreatitis , Aged , Humans , Middle Aged , Acute Disease , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Fluid Therapy/methods , Network Meta-Analysis , Pancreatitis/prevention & control , Pancreatitis/etiology , Ringer's Lactate/therapeutic use , Ringer's Lactate/administration & dosage , Risk Factors , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Treatment Outcome
17.
Cell Rep ; 43(5): 114197, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38733587

ABSTRACT

Interneurons (INs), specifically those in disinhibitory circuits like somatostatin (SST) and vasoactive intestinal peptide (VIP)-INs, are strongly modulated by the behavioral context. Yet, the mechanisms by which these INs are recruited during active states and whether their activity is consistent across sensory cortices remain unclear. We now report that in mice, locomotor activity strongly recruits SST-INs in the primary somatosensory (S1) but not the visual (V1) cortex. This diverse engagement of SST-INs cannot be explained by differences in VIP-IN function but is absent in the presence of visual input, suggesting the involvement of feedforward sensory pathways. Accordingly, inactivating the somatosensory thalamus, but not decreasing VIP-IN activity, significantly reduces the modulation of SST-INs by locomotion. Model simulations suggest that the differences in SST-INs across behavioral states can be explained by varying ratios of VIP- and thalamus-driven activity. By integrating feedforward activity with neuromodulation, SST-INs are anticipated to be crucial for adapting sensory processing to behavioral states.


Subject(s)
Interneurons , Somatostatin , Vasoactive Intestinal Peptide , Animals , Interneurons/metabolism , Interneurons/physiology , Somatostatin/metabolism , Mice , Vasoactive Intestinal Peptide/metabolism , Somatosensory Cortex/physiology , Somatosensory Cortex/metabolism , Male , Mice, Inbred C57BL , Locomotion/physiology , Behavior, Animal/physiology , Visual Cortex/physiology , Visual Cortex/metabolism , Thalamus/physiology , Thalamus/metabolism
18.
Eur J Endocrinol ; 190(6): 421-433, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38701338

ABSTRACT

INTRODUCTION: Growth hormone (GH)-secreting pituitary tumors (GHomas) are the most common acromegaly cause. At diagnosis, most of them are macroadenomas, and up to 56% display cavernous sinus invasion. Biomarker assessment associated with tumor growth and invasion is important to optimize their management. OBJECTIVES: The study aims to identify clinical/hormonal/molecular biomarkers associated with tumor size and invasiveness in GHomas and to analyze the influence of pre-treatment with somatostatin analogs (SSAs) or dopamine agonists (DAs) in key molecular biomarker expression. METHODS: Clinical/analytical/radiological variables were evaluated in 192 patients from the REMAH study (ambispective multicenter post-surgery study of the Spanish Society of Endocrinology and Nutrition). The expression of somatostatin/ghrelin/dopamine system components and key pituitary/proliferation markers was evaluated in GHomas after the first surgery. Univariate/multivariate regression studies were performed to identify association between variables. RESULTS: Eighty percent of patients harbor macroadenomas (63.8% with extrasellar growth). Associations between larger and more invasive GHomas with younger age, visual abnormalities, higher IGF1 levels, extrasellar/suprasellar growth, and/or cavernous sinus invasion were found. Higher GH1 and lower PRL/POMC/CGA/AVPR1B/DRD2T/DRD2L expression levels (P < .05) were associated with tumor invasiveness. Least Absolute Shrinkage and Selection Operator's penalized regression identified combinations of clinical and molecular features with areas under the curve between 0.67 and 0.82. Pre-operative therapy with DA or SSAs did not alter the expression of any of the markers analyzed except for DRD1/AVPR1B (up-regulated with DA) and FSHB/CRHR1 (down-regulated with SSAs). CONCLUSIONS: A specific combination of clinical/analytical/molecular variables was found to be associated with tumor invasiveness and growth capacity in GHomas. Pre-treatment with first-line drugs for acromegaly did not significantly modify the expression of the most relevant biomarkers in our association model. These findings provide valuable insights for risk stratification and personalized management of GHomas.


Subject(s)
Acromegaly , Adenoma , Growth Hormone-Secreting Pituitary Adenoma , Neoplasm Invasiveness , Humans , Male , Female , Acromegaly/metabolism , Middle Aged , Adult , Growth Hormone-Secreting Pituitary Adenoma/pathology , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Adenoma/metabolism , Adenoma/pathology , Aged , Dopamine Agonists/therapeutic use , Biomarkers, Tumor/metabolism , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Human Growth Hormone/metabolism
19.
Clin Nucl Med ; 49(7): 695-697, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38768160

ABSTRACT

ABSTRACT: 64 Cu-DOTATATE PET/CT of a 44-year-old man with an ileal neuroendocrine tumor demonstrated the primary tumor, local nodal metastases, and a pericaval nodal metastasis. Localization of the pericaval node during surgery may be difficult, thus 4.4 mCi of 111 In-pentetreotide was administered before surgery to assist with localization and resection. At surgery, the pericaval nodal metastasis was readily detected by gamma probe, which could then be resected and pathologically proven to be a metastasis. This demonstrates the use of somatostatin receptor-targeted imaging for intraoperative localization of an otherwise difficult to surgically localize metastasis. Without intraoperative somatostatin receptor-targeted radiosurgery, disease may have been incompletely resected.


Subject(s)
Ileal Neoplasms , Neuroendocrine Tumors , Radiosurgery , Somatostatin , Humans , Male , Adult , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/surgery , Neuroendocrine Tumors/pathology , Ileal Neoplasms/diagnostic imaging , Ileal Neoplasms/surgery , Ileal Neoplasms/pathology , Somatostatin/analogs & derivatives , Lymphatic Metastasis , Retroperitoneal Neoplasms/diagnostic imaging , Retroperitoneal Neoplasms/surgery , Retroperitoneal Neoplasms/pathology , Intraoperative Period , Surgery, Computer-Assisted , Positron Emission Tomography Computed Tomography
20.
Endocr Relat Cancer ; 31(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38713182

ABSTRACT

The objective of the study was to evaluate the efficacy of second-line therapies in patients with acromegaly caused by a growth hormone (GH) and prolactin (PRL) co-secreting pituitary neuroendocrine tumor (GH&PRL-Pit-NET) compared to their efficacy in patients with acromegaly caused by a GH-secreting pituitary neuroendocrine tumor (GH-Pit-NET). This is a multicenter retrospective study of patients with acromegaly on treatment with pasireotide and/or pegvisomant. Patients were classified in two groups: GH&PRL-Pit-NETs when evidence of hyperprolactinemia and immunohistochemistry (IHC) for GH and PRL was positive or if PRL were >200 ng/dL regardless of the PRL-IHC and GH-Pit-NETs when the previously mentioned criteria were not met. A total of 28 cases with GH&PRL-Pit-NETs and 122 with GH-Pit-NETs met the inclusion criteria. GH&PRL-Pit-NETs presented at a younger age, caused hypopituitarism, and were invasive more frequently than GH-Pit-NETs. There were 124 patients treated with pegvisomant and 49 with pasireotide at any time. The efficacy of pegvisomant for IGF-1 normalization was of 81.5% and of pasireotide of 71.4%. No differences in IGF-1 control with pasireotide and with pegvisomant were observed between GH&PRL-Pit-NETs and GH-Pit-NETs. All GH&PRL-Pit-NET cases treated with pasireotide (n = 6) and 82.6% (n = 19/23) of the cases treated with pegvisomant normalized PRL levels. No differences in the rate of IGF-1 control between pegvisomant and pasireotide were detected in patients with GH&PRL-Pit-NETs (84.9% vs 66.7%, P = 0.178). We conclude that despite the more aggressive behavior of GH&PRL-Pit-NETs than GH-Pit-NETs, no differences in the rate of IGF-1 control with pegvisomant and pasireotide were observed between both groups, and both drugs have shown to be effective treatments to control IGF-1 and PRL hypersecretion in these tumors.


Subject(s)
Acromegaly , Human Growth Hormone , Neuroendocrine Tumors , Prolactin , Somatostatin , Humans , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Male , Female , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/therapeutic use , Middle Aged , Adult , Prolactin/blood , Prolactin/metabolism , Retrospective Studies , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Acromegaly/drug therapy , Acromegaly/metabolism , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...