Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 56
1.
Nucl Med Biol ; 118-119: 108329, 2023.
Article En | MEDLINE | ID: mdl-36805869

Neurotensin receptor 1 (NTSR1) can stimulate tumor proliferation through neurotensin (NTS) activation and are overexpressed by a variety of cancers. The high binding affinity of NTS/NTSR1 makes radiolabeled NTS derivatives interesting for cancer diagnosis and staging. Internalization of NTS/NTSR1 also suggests therapeutic application with high LET alpha particles and low energy electrons. We investigated the therapeutic efficacy of [58mCo]Co-NOTA-NT-20.3 in vivo using murine models xenografted with NTSR1-positive HT29 human colorectal adenocarcinoma cells, and utilized [55Co]Co-NOTA-NT-20.3 for dosimetry. METHODS: Targeting properties and cytotoxicity of [55/58mCo]Co-NOTA-NT-20.3 were assessed with HT29 cells. Female nude mice were xenografted with HT29 tumors and administered [55Co or 58mCo]Co-NOTA-NT-20.3 to evaluate pharmacokinetics or for therapy, respectively. Dosimetry calculations followed the Medical Internal Radiation Dose (MIRD) formalism and human absorbed dose rate per unit activity were obtained from OpenDose. The pilot therapy study consisted of two groups (each N = 3) receiving 110 ± 15 MBq and 26 ± 6 MBq [58mCo]Co-NOTA-NT-20.3 one week after tumor inoculation, and control (N = 3). Tumor sizes and masses were measured twice a week after therapy. Complete blood count and kidney histology were also performed to assess toxicity. RESULTS: HPLC measured radiochemical purity of [55,58mCo]Co-NOTA-NT-20.3 > 99 %. Labeled compounds retained NTS targeting properties. [58mCo]Co-NOTA-NT-20.3 exhibited cytotoxicity for HT29 cells and was >15× more potent than [58mCo]CoCl2. Xenografted tumors responded modestly to administered doses, but mice showed no signs of radiotoxicity. Absorbed dose to tumor and kidney with 110 MBq [58mCo]Co-NOTA-NT-20.3 were 0.6 Gy and 0.8 Gy, respectively, and other organs received less than half of the absorbed dose to tumor. Off-target radiation dose from cobalt-58g was small but reduces the therapeutic window. CONCLUSION: The enhanced in vitro cytotoxicity and high tumor-to-background led us to investigate the therapeutic efficacy of [58mCo]Co-NOTA-NT-20.3 in vivo. Although we were unable to induce tumor response commensurate with [177Lu]Lu-NT127 (NLys-Lys-Pro-Tyr-Tle-Leu) studies involving similar time-integrated activity, the absence of observed toxicity may constitute an opportunity for targeting vectors with improved uptake and/or retention to avoid the aftereffects of other high-LET radioactive emissions. Future studies with higher uptake, activity and/or multiple dosing regimens are warranted. The theranostic approach employed in this work was crucial for dosimetry analysis.


Precision Medicine , Receptors, Neurotensin , Female , Mice , Humans , Animals , Receptors, Neurotensin/metabolism , Pilot Projects , Mice, Nude , Neurotensin/therapeutic use , Neurotensin/metabolism
2.
Nord J Psychiatry ; 76(7): 559-564, 2022 Oct.
Article En | MEDLINE | ID: mdl-36189960

INTRODUCTION: Nuclear distribution element like-1 (Ndel1) is a cytosolic oligopeptidase, which was suggested as a potential biomarker of aberrant neurodevelopment and early stage of schizophrenia (SCZ). The involvement of Ndel1 in neurite outgrowth, neuronal migration and neurodevelopment was demonstrated. Moreover, Ndel1 cleaves neuropeptides, including the endogenous antipsychotic peptide neurotensin, and lower Ndel1 activity was reported in SCZ patients compared with healthy controls (HCs). Changes in brain-derived neurotrophic factor (BDNF) and inflammatory cytokines levels were also implicated in SCZ. OBJECTIVE: This preliminary study aimed to investigate the interactions between these immune and neurodevelopmental/neurotrophic biomarkers, namely BDNF and the recently identified SCZ biomarker Ndel1. RESULTS: We observed lower Ndel1 activity and IL-4 levels, and higher BDNF levels, in plasma of SCZ (N = 23) compared with HCs (N = 29). Interestingly, significant correlation between Ndel1 activity and IL-4 levels was observed in SCZ, while no correlation with any other evaluated interleukins (namely IL-2, IL-8, IL-10 and IL-17A) or BDNF levels was noticed. CONCLUSION: Although this hypothesis needs to be further explored for a better understanding of the mechanisms by which these altered pathways are associated to each other in SCZ, we suggest that Ndel1 and the inflammatory marker IL-4 are directly correlated.


Antipsychotic Agents , Neuropeptides , Schizophrenia , Antipsychotic Agents/therapeutic use , Biomarkers , Brain-Derived Neurotrophic Factor , Cytokines , Humans , Interleukin-10/therapeutic use , Interleukin-17/therapeutic use , Interleukin-2/therapeutic use , Interleukin-4/therapeutic use , Interleukin-8/therapeutic use , Neurotensin/therapeutic use , Peptides/therapeutic use , Schizophrenia/drug therapy
3.
J Med Chem ; 64(4): 2110-2124, 2021 02 25.
Article En | MEDLINE | ID: mdl-33538583

Neurotensin (NT) receptor type 2 (NTS2) represents an attractive target for the development of new NT-based analgesics. Here, we report the synthesis and functional in vivo characterization of the first constrained NTS2-selective macrocyclic NT analog. While most chemical optimization studies rely on the NT(8-13) fragment, we focused on NT(7-12) as a scaffold to design NTS2-selective macrocyclic peptides. Replacement of Ile12 by Leu, and Pro7/Pro10 by allylglycine residues followed by cyclization via ring-closing metathesis led to macrocycle 4, which exhibits good affinity for NTS2 (50 nM), high selectivity over NTS1 (>100 µM), and improved stability compared to NT(8-13). In vivo profiling in rats reveals that macrocycle 4 produces potent analgesia in three distinct rodent pain models, without causing the undesired effects associated with NTS1 activation. We further provide evidence of its non-opioid antinociceptive activity, therefore highlighting the strong therapeutic potential of NTS2-selective analogs for the management of acute and chronic pain.


Analgesics/therapeutic use , Neurotensin/analogs & derivatives , Neurotensin/therapeutic use , Pain/drug therapy , Peptides, Cyclic/therapeutic use , Receptors, Neurotensin/metabolism , Analgesics/chemical synthesis , Animals , Drug Design , Male , Molecular Structure , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , Peptides, Cyclic/chemical synthesis , Rats, Sprague-Dawley , Structure-Activity Relationship
4.
Future Med Chem ; 12(19): 1779-1803, 2020 10.
Article En | MEDLINE | ID: mdl-33032465

The natural tridecapeptide neurotensin has been emerged as a promising therapeutic scaffold for the treatment of neurological diseases and cancer. In this work, we aimed to identify the top 100 most cited original research papers as well as recent key studies related to neurotensins. The Web of Science Core Collection database was searched and the retrieved research articles were analyzed by using the VOSviewer software. The most cited original articles were published between 1973 and 2013. The top-cited article was by Carraway and Leeman reporting the discovery of neurotensin in 1973. The highly cited terms were associated with hypotension and angiotensin-converting-enzyme. The conducted analysis reveals the therapeutic potentials of neurotensin, and further impactful research toward its clinical development is warrantied.


Antineoplastic Agents/therapeutic use , Central Nervous System Agents/therapeutic use , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Neurotensin/therapeutic use , Animals , Antineoplastic Agents/chemistry , Central Nervous System Agents/chemistry , Humans , Neurotensin/chemistry
5.
Eur J Pharmacol ; 882: 173174, 2020 Sep 05.
Article En | MEDLINE | ID: mdl-32534076

Neurotensin (NT) exerts naloxone-insensitive antinociceptive action through its binding to both NTS1 and NTS2 receptors and NT analogs provide stronger pain relief than morphine on a molecular basis. Here, we examined the analgesic/adverse effect profile of a new NT(8-13) derivative denoted JMV2009, in which the Pro10 residue was substituted by a silicon-containing unnatural amino acid silaproline. We first report the synthesis and in vitro characterization (receptor-binding affinity, functional activity and stability) of JMV2009. We next examined its analgesic activity in a battery of acute, tonic and chronic pain models. We finally evaluated its ability to induce adverse effects associated with chronic opioid use, such as constipation and analgesic tolerance or related to NTS1 activation, like hypothermia. In in vitro assays, JMV2009 exhibited high binding affinity for both NTS1 and NTS2, improved proteolytic resistance as well as agonistic activities similar to NT, inducing sustained activation of p42/p44 MAPK and receptor internalization. Intrathecal injection of JMV2009 produced dose-dependent antinociceptive responses in the tail-flick test and almost completely abolished the nociceptive-related behaviors induced by chemical somatic and visceral noxious stimuli. Likewise, increasing doses of JMV2009 significantly reduced tactile allodynia and weight bearing deficits in nerve-injured rats. Importantly, repeated agonist treatment did not result in the development of analgesic tolerance. Furthermore, JMV2009 did not cause constipation and was ineffective in inducing hypothermia. These findings suggest that NT drugs can act as an effective opioid-free medication for the management of pain or can serve as adjuvant analgesics to reduce the opioid adverse effects.


Analgesics/therapeutic use , Neurotensin/analogs & derivatives , Neurotensin/therapeutic use , Pain/drug therapy , Receptors, Neurotensin/agonists , Analgesics/pharmacology , Animals , Blood Pressure/drug effects , Body Temperature/drug effects , Gastrointestinal Motility/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Neurotensin/pharmacology , Pain/physiopathology , Rats, Sprague-Dawley , Receptors, Neurotensin/physiology
6.
Nucl Med Commun ; 41(5): 411-415, 2020 May.
Article En | MEDLINE | ID: mdl-32168264

Pancreatic cancer is the fourth leading cause of cancer-related death in both men and women. Neurotensin receptors are overexpressed in different malignancies, above all pancreatic cancer. On the other hand, neurotensin receptor expression in inflammation is quite low. This fact can probably solve the most important problem of F-FDG PET imaging - distinguishing malignant and inflammatory processes. The first therapeutic injection of radiolabelled neurotensin in human with pancreatic cancer has been successfully performed. Animal experiments are also very close to the first in human injection of radiolabelled neurotensin for diagnostic purposes. The purpose of this article is to provide an overview of radiolabelled neurotensin analogues that can be used in imaging and therapy in patients with pancreatic ductal adenocarcinoma.


Neurotensin/therapeutic use , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/radiotherapy , Positron-Emission Tomography/methods , Animals , Humans , Isotope Labeling , Neurotensin/analogs & derivatives , Neurotensin/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Neurotensin/metabolism
7.
Anticancer Agents Med Chem ; 20(4): 402-416, 2020.
Article En | MEDLINE | ID: mdl-31889492

BACKGROUND: Cancer is the leading cause of death worldwide. Early detection can reduce the disadvantageous effects of diseases and the mortality in cancer. Nuclear medicine is a powerful tool that has the ability to diagnose malignancy without harming normal tissues. In recent years, radiolabeled peptides have been investigated as potent agents for cancer detection. Therefore, it is necessary to modify radiopeptides in order to achieve more effective agents. OBJECTIVE: This review describes modifications in the structure of radioconjugates with spacers who have improved the specificity and sensitivity of the peptides that are used in oncologic diagnosis and therapy. METHODS: To improve the biological activity, researchers have conjugated these peptide analogs to different spacers and bifunctional chelators. Many spacers of different kinds, such as hydrocarbon chain, amino acid sequence, and poly (ethyleneglycol) were introduced in order to modify the pharmacokinetic properties of these biomolecules. RESULTS: Different spacers have been applied to develop radiolabeled peptides as potential tracers in nuclear medicine. Spacers with different charge and hydrophilicity affect the characteristics of peptide conjugate. For example, the complex with uncharged and hydrophobic spacers leads to increased liver uptake, while the composition with positively charged spacers results in high kidney retention. Therefore, the pharmacokinetics of radio complexes correlates to the structure and total charge of the conjugates. CONCLUSION: Radio imaging technology has been successfully applied to detect a tumor in the earliest stage. For this purpose, the assessment of useful agents to diagnose the lesion is necessary. Developing peptide radiopharmaceuticals using spacers can improve in vitro and in vivo behavior of radiotracers leading to better noninvasive detection and monitoring of tumor growth.


Neoplasms/diagnosis , Neoplasms/radiotherapy , Peptides/chemistry , Peptides/therapeutic use , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Animals , Bombesin/chemistry , Bombesin/therapeutic use , Gastrins/chemistry , Gastrins/therapeutic use , Humans , Melanocyte-Stimulating Hormones/chemistry , Melanocyte-Stimulating Hormones/therapeutic use , Neurotensin/chemistry , Neurotensin/therapeutic use , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Somatostatin/chemistry , Somatostatin/therapeutic use
8.
Diabetes Metab Res Rev ; 34(6): e3006, 2018 09.
Article En | MEDLINE | ID: mdl-29633491

Xenin-25 is a 25-amino acid peptide hormone co-secreted from the same enteroendocrine K-cell as the incretin peptide glucose-dependent insulinotropic polypeptide. There is no known specific receptor for xenin-25, but studies suggest that at least some biological actions may be mediated through interaction with the neurotensin receptor. Original investigation into the physiological significance of xenin-25 focussed on effects related to gastrointestinal transit and satiety. However, xenin-25 has been demonstrated in pancreatic islets and recently shown to possess actions in relation to the regulation of insulin and glucagon secretion, as well as promoting beta-cell survival. Accordingly, the beneficial impact of xenin-25, and related analogues, has been assessed in animal models of diabetes-obesity. In addition, studies have demonstrated that metabolically active fragment peptides of xenin-25, particularly xenin-8, possess independent therapeutic promise for diabetes, as well as serving as bioactive components for the generation of multi-acting hybrid peptides with antidiabetic potential. This review focuses on continuing developments with xenin compounds in relation to new therapeutic approaches for diabetes-obesity.


Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Neurotensin/therapeutic use , Obesity/drug therapy , Therapies, Investigational/trends , Animals , Humans , Peptide Fragments/therapeutic use , Peptides/therapeutic use , Therapies, Investigational/methods
9.
Diabetes Obes Metab ; 20(5): 1166-1175, 2018 05.
Article En | MEDLINE | ID: mdl-29316242

AIMS: To demarcate pathological events in the brain as a result of short-term to chronic high-fat-diet (HFD) feeding, which leads to cognitive impairment and neuroinflammation, and to assess the efficacy of Xenin-25[Lys(13)PAL] in chronic HFD-fed mice. METHODS: C57BL/6 mice were fed an HFD or a normal diet for 18 days, 34 days, 10 and 21 weeks. Cognition was assessed using novel object recognition and the Morris water maze. Markers of insulin signalling and inflammation were measured in brain and plasma using immunohistochemistry, quantitative PCR and multi-array technology. Xenin-25[Lys(13)PAL] was also administered for 5 weeks in chronic HFD-fed mice to assess therapeutic potential at a pathological stage. RESULTS: Recognition memory was consistently impaired in HFD-fed mice and spatial learning was impaired in 18-day and 21-week HFD-fed mice. Gliosis, oxidative stress and IRS-1 pSer616 were increased in the brain on day 18 in HFD-fed mice and were reduced by Xenin-25[Lys(13)PAL] in 21-week HFD-fed mice. In plasma, HFD feeding elevated interleukin (IL)-6 and chemokine (C-X-C motif) ligand 1 at day 34 and IL-5 at week 10. In the brain, HFD feeding reduced extracellular signal-regulated kinase 2 (ERK2), mechanistic target of rapamycin (mTOR), NF-κB1, protein kinase C (PKC)θ and Toll-like receptor 4 (TLR4) mRNA at week 10 and increased expression of glucacon-like peptide-1 receptor, inhibitor of NF-κB kinase ß, ERK2, mTOR, NF-κB1, PKCθ and TLR4 at week 21, elevations that were abrogated by Xenin-25[Lys(13)PAL]. CONCLUSIONS: HFD feeding modulates cognitive function, synapse density, inflammation and insulin resistance in the brain. Xenin-25[Lys(13)PAL] ameliorated markers of inflammation and insulin signalling dysregulation and may have therapeutic potential in the treatment of diseases associated with neuroinflammation or perturbed insulin signalling in the brain.


Brain/drug effects , Cognition Disorders/drug therapy , Disease Models, Animal , Encephalitis/drug therapy , Insulin Resistance , Neurotensin/analogs & derivatives , Nootropic Agents/therapeutic use , Peptides/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Behavior, Animal/drug effects , Biomarkers/blood , Biomarkers/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Cognition Disorders/immunology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Diet, High-Fat/adverse effects , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/pathology , Exploratory Behavior/drug effects , Gene Expression Regulation, Developmental/drug effects , Immunohistochemistry , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neurotensin/therapeutic use , Oxidative Stress/drug effects , Random Allocation
10.
Diabetologia ; 60(3): 541-552, 2017 03.
Article En | MEDLINE | ID: mdl-28004148

AIMS/HYPOTHESIS: Glucose-dependent insulinotropic polypeptide (GIP) and xenin, regulatory gut hormones secreted from enteroendocrine K cells, exert important effects on metabolism. In addition, xenin potentiates the biological actions of GIP. The present study assessed the actions and therapeutic utility of a (DAla2)GIP/xenin-8-Gln hybrid peptide, in comparison with the parent peptides (DAla2)GIP and xenin-8-Gln. METHODS: Following confirmation of enzymatic stability, insulin secretory activity of (DAla2)GIP/xenin-8-Gln was assessed in BRIN-BD11 beta cells. Acute and persistent glucose-lowering and insulin-releasing effects were then examined in vivo. Finally, the metabolic benefits of twice daily injection of (DAla2)GIP/xenin-8-Gln was determined in high-fat-fed mice. RESULTS: All peptides significantly (p < 0.05 to p < 0.001) enhanced in vitro insulin secretion from pancreatic clonal BRIN-BD11 cells, with xenin (and particularly GIP)-related signalling pathways, being important for this action. Administration of (DAla2)GIP or (DAla2)GIP/xenin-8-Gln in combination with glucose significantly (p < 0.05) lowered blood glucose and increased plasma insulin in mice, with a protracted response of up to 4 h. All treatments elicited appetite-suppressive effects (p < 0.05), particularly (DAla2)GIP/xenin-8-Gln and xenin-8-Gln at elevated doses of 250 nmol/kg. Twice-daily administration of (DAla2)GIP/xenin-8-Gln or (DAla2)GIP for 21 days to high-fat-fed mice returned circulating blood glucose to lean control levels. In addition, (DAla2)GIP/xenin-8-Gln treatment significantly (p < 0.05) reduced glycaemic levels during a 24 h glucose profile assessment. Neither of the treatment regimens had an effect on body weight, energy intake or circulating insulin concentrations. However, insulin sensitivity was significantly (p < 0.001) improved by both treatments. Interestingly, GIP-mediated glucose-lowering (p < 0.05) and insulin-releasing (p < 0.05 to p < 0.01) effects were substantially improved by (DAla2)GIP and (DAla2)GIP/xenin-8-Gln treatment. Pancreatic islet and beta cell area (p < 0.001), as well as pancreatic insulin content (p < 0.05), were augmented in (DAla2)GIP/xenin-8-Gln-treated mice, related to enhanced proliferation and decreased apoptosis of beta cells, whereas (DAla2)GIP evoked increases (p < 0.05 to p < 0.01) in islet number. CONCLUSIONS/INTERPRETATION: These studies highlight the clear potential of GIP/xenin hybrids for the treatment of type 2 diabetes.


Gastric Inhibitory Polypeptide/therapeutic use , Neurotensin/therapeutic use , Peptides/therapeutic use , Animals , Blood Glucose/drug effects , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Glucose/metabolism , Homeostasis/drug effects , Immunohistochemistry , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Peptide Fragments/therapeutic use
11.
PLoS One ; 11(3): e0152818, 2016.
Article En | MEDLINE | ID: mdl-27032106

Xenin is a peptide that is co-secreted with the incretin hormone, glucose-dependent insulinotropic polypeptide (GIP), from intestinal K-cells in response to feeding. Studies demonstrate that xenin has appetite suppressive effects and modulates glucose-induced insulin secretion. The present study was undertaken to determine the bioactivity and antidiabetic properties of two C-terminal fragment xenin peptides, namely xenin 18-25 and xenin 18-25 Gln. In BRIN-BD11 cells, both xenin fragment peptides concentration-dependently stimulated insulin secretion, with similar efficacy as the parent peptide. Neither fragment peptide had any effect on acute feeding behaviour at elevated doses of 500 nmol/kg bw. When administered together with glucose to normal mice at 25 nmol/kg bw, the overall insulin secretory effect was significantly enhanced in both xenin 18-25 and xenin 18-25 Gln treated mice, with better moderation of blood glucose levels. Twice daily administration of xenin 18-25 or xenin 18-25 Gln for 21 days in high fat fed mice did not affect energy intake, body weight, circulating blood glucose or body fat stores. However, circulating plasma insulin concentrations had a tendency to be elevated, particularly in xenin 18-25 Gln mice. Both treatment regimens significantly improved insulin sensitivity by the end of the treatment period. In addition, sustained treatment with xenin 18-25 Gln significantly reduced the overall glycaemic excursion and augmented the insulinotropic response to an exogenous glucose challenge on day 21. In harmony with this, GIP-mediated glucose-lowering and insulin-releasing effects were substantially improved by twice daily xenin 18-25 Gln treatment. Overall, these data provide evidence that C-terminal octapeptide fragments of xenin, such as xenin 18-25 Gln, have potential therapeutic utility for type 2 diabetes.


Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Hormones/therapeutic use , Hypoglycemic Agents/therapeutic use , Neurotensin/therapeutic use , Amino Acid Sequence , Animals , Blood Glucose/analysis , Body Weight/drug effects , Diabetes Mellitus, Type 2/blood , Diet, High-Fat , Energy Intake/drug effects , Gastrointestinal Hormones/chemistry , Glucose Tolerance Test , Hypoglycemic Agents/chemistry , Insulin/blood , Insulin Resistance , Male , Mice , Molecular Sequence Data , Neurotensin/chemistry
12.
Behav Brain Res ; 305: 8-17, 2016 May 15.
Article En | MEDLINE | ID: mdl-26909848

Although neurotensin (NT) analogs are known to produce antipsychotic-like effects, the therapeutic possibility of a brain penetrant NTS1 agonist in treating psychiatric disorders has not been well studied. Here, we examined whether PD149163, a brain-penetrant NTS1-specific agonist, displays antipsychotic-like effects in C57BL/6J mice by investigating the effect of PD149163 on amphetamine-mediated hyperactivity and amphetamine-induced disruption of prepulse inhibition. In addition, we assessed the effect of PD149163 on glycogen synthase kinase-3 (GSK-3) activity, a downstream molecular target of antipsychotics and mood stabilizers, using phospho-specific antibodies. PD149163 (0.1 and 0.5mg/kg) inhibited amphetamine-induced hyperactivity in mice, indicating that NTS1 activation inhibits psychomotor agitation. PD149163 (0.5mg/kg) also increased prepulse inhibition, suggesting that NTS1 activation reduces prepulse inhibition deficits which often co-occur with psychosis in humans. Interestingly, PD149163 increased the inhibitory serine phosphorylation on both GSK-3α and GSK-3ß in a dose- and time-dependent manner in the nucleus accumbens and medial prefrontal cortex of the mice. Moreover, PD149163 inhibited GSK-3 activity in the nucleus accumbens and medial prefrontal cortex in the presence of amphetamine. Thus, like most current antipsychotics and mood stabilizers, PD149163 inhibited GSK-3 activity in cortico-striatal circuitry. Together, our findings indicate that PD149163 may be a novel antipsychotic.


Antipsychotic Agents/therapeutic use , Neurotensin/analogs & derivatives , Psychomotor Agitation/drug therapy , Amphetamine/toxicity , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Glycogen Synthase Kinase 3/metabolism , Male , Mice , Mice, Inbred C57BL , Neurotensin/therapeutic use , Phosphorylation/drug effects , Prepulse Inhibition/drug effects , Psychomotor Agitation/etiology , Serine/metabolism , Time Factors
13.
Biomed Res Int ; 2015: 173507, 2015.
Article En | MEDLINE | ID: mdl-25984525

Despite recent advances in multimodal therapy, bladder cancer still ranks ninth in worldwide cancer incidence. New molecules which might improve early diagnosis and therapeutic efficiency for tumors of such high epidemiological impact therefore have very high priority. In the present study, the tetrabranched neurotensin peptide NT4 was conjugated with functional units for cancer-cell imaging or therapy and was tested on bladder cancer cell lines and specimens from bladder cancer surgical resections, in order to evaluate its potential for targeted personalized therapy of bladder cancer. Fluorophore-conjugated NT4 distinguished healthy and cancer tissues with good statistical significance (P < 0.05). NT4 conjugated to methotrexate or gemcitabine was cytotoxic for human bladder cancer cell lines at micromolar concentrations. Their selectivity for bladder cancer tissue and capacity to carry tracers or drugs make NT4 peptides candidate tumor targeting agents for tracing cancer cells and for personalized therapy of human bladder cancer.


Neurotensin/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Biopsy , Cell Death/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Endocytosis/drug effects , Female , Fluorescence , Humans , Male , Methotrexate/pharmacology , Middle Aged , Neurotensin/pharmacology , Protein Binding/drug effects , Urinary Bladder Neoplasms/pathology , Gemcitabine
14.
Curr Pharm Des ; 21(7): 840-8, 2015.
Article En | MEDLINE | ID: mdl-25345606

Neurotensin (NT) is an endogenous 13 amino acid neuropeptide with profound opioid-independent analgesic effects. This role of NT is thought to be mediated by both neurotensin receptor subtype 1 (NTS1) and neurotensin receptor subtype 2 (NTS2). NT and its receptors are widely distributed in the pain circuits in central nervous system. Thus NT might modulate pain in many structures of pain pathway, such as spinal cord, rostroventral medulla (RVM) and periaqueductal gray (PAG). Actually either intrathecal application of NT or direct injection of NT into RVM or PAG or intracerebroventricular injection of NT showed analgesic effects. NT exerted its antinociceptive effects in both acute pain and chronic pain models. The analgesic effects of NT were originally found in acute pain experiments. In the case of pathological pain, for example, formalin injection induced inflammatory pain and sciatic nerve constriction induced neuropathic pain, NT also shows antinociceptive effects. The effects exist in somatic pain as well as visceral pain induced by noxious colorectal distension (CRD) or writhing test. It should be noted that NT plays an important role in stress-induced antinociception (SIAN), especially in higher intensity stress experiments. However as a neuropeptide, NT is susceptible to degradation by peptidases and cannot cross the blood-brain barrier (BBB). Great efforts have been made to find NT analogues that are more biologically stable and could inhibit pain by systematic administration. The present review focuses on the analgesic role and the underlying mechanisms of NT and its analogues in pain, especially in chronic pain models.


Analgesics, Non-Narcotic/therapeutic use , Neurotensin/analogs & derivatives , Neurotensin/therapeutic use , Pain/drug therapy , Pain/metabolism , Analgesics, Opioid/therapeutic use , Animals , Humans
15.
Exp Clin Psychopharmacol ; 22(6): 548-56, 2014 Dec.
Article En | MEDLINE | ID: mdl-25285844

Major depressive disorder is the most common mood disorder in the United States and European Union; however, the limitations of clinically available antidepressant drugs have led researchers to pursue novel pharmacological treatments. Clinical studies have reported that monotherapy with the atypical antipsychotic drug quetiapine produces a rapid reduction in depressive symptoms that is apparent after 1 week of treatment, and it is possible that the active metabolite N-desalkylquetiapine, which structurally resembles an antidepressant drug, produces antidepressant effects. Neuropharmacological evaluations of the neurotensin NTS1 receptor agonist PD149163 suggest antidepressant efficacy, but the effects of a NTS1 receptor agonist in an antidepressant animal model have yet to be reported. The present study examined the antidepressant-like effects of N-desalkylquetiapine, PD14916, quetiapine, the tricyclic antidepressant drug imipramine, the atypical antipsychotic drug risperidone, and the typical antipsychotic drug raclopride on responding in male Sprague-Dawley rats trained on a differential-reinforcement-of-low-rate 72-s operant schedule, a procedure used for screening antidepressant drugs. Quetiapine, PD149163, risperidone, and imipramine exhibited antidepressant-like effects by increasing the number of reinforcers earned, decreasing the number of responses emitted, and shifting the interresponse time (IRT) distributions to the right. N-Desalkylquetiapine produced a partial antidepressant-like effect by decreasing the number of responses emitted and producing a rightward shift in the IRT distributions, but it did not significantly alter the number of reinforcers earned. Raclopride decreased reinforcers and responses. These data suggest that N-desalkylquetiapine likely contributes to quetiapine's antidepressant efficacy and identify NTS1 receptor activation as a potential novel pharmacologic strategy for antidepressant drugs.


Antidepressive Agents, Second-Generation/therapeutic use , Antipsychotic Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Dibenzothiazepines/chemistry , Disease Models, Animal , Neurotensin/analogs & derivatives , Receptors, Neurotensin/agonists , Alkylation , Animals , Antidepressive Agents/therapeutic use , Antidepressive Agents, Tricyclic/therapeutic use , Behavior, Animal/drug effects , Conditioning, Operant/drug effects , Dibenzothiazepines/therapeutic use , Male , Neurotensin/therapeutic use , Quetiapine Fumarate , Rats, Sprague-Dawley , Reinforcement, Psychology
16.
Exp Clin Psychopharmacol ; 22(6): 541-7, 2014 Dec.
Article En | MEDLINE | ID: mdl-25222546

Agonists for the neurotensin NTS1 receptor consistently exhibit antipsychotic effects in animal models without producing catalepsy, suggesting that NTS1-receptor agonists may be a novel class of drugs to treat schizophrenia. Moreover, studies utilizing NTS1 agonists have reported improvements in some aspects of cognitive functioning, including prepulse inhibition and learning procedures, which suggest an ability of NTS1-receptor agonists to diminish neurocognitive deficits. The present study sought to assess both baseline delay-induced memory performance and the effects of NTS1-receptor activation on learning and memory consolidation in male Long-Evans and Brown Norway rats using a delayed nonmatch-to-position task radial arm-maze task. In the absence of drugs, Brown Norway rats displayed a significant increase in spatial memory errors following 3-, 7-, and 24-hr delay, whereas Long-Evans rats exhibited an increase in spatial memory errors following only a 7-, and 24-hr delay. With Brown Norway rats, administration of PD149163 before or after an information trial significantly reduced errors during a retention trial after a 24 hr delay. Administration of the NTS(1/2)-receptor antagonist SR142948 prior to the information trial did not affect retention-trial errors. These data are consistent with previous findings that Brown Norway rats have natural cognitive deficits and that they may be useful for assessing putative antipsychotic drugs for cognitive efficacy. Moreover, the results of this study support previous findings suggesting that NTS1-receptor agonists may improve some aspects of cognitive functioning.


Antipsychotic Agents/therapeutic use , Disease Models, Animal , Memory Disorders/drug therapy , Neurotensin/analogs & derivatives , Nootropic Agents/therapeutic use , Receptors, Neurotensin/agonists , Animals , Antipsychotic Agents/administration & dosage , Behavior, Animal/drug effects , Cognition/drug effects , Dose-Response Relationship, Drug , Injections, Subcutaneous , Male , Maze Learning/drug effects , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Neurotensin/administration & dosage , Neurotensin/therapeutic use , Nootropic Agents/administration & dosage , Pyrazoles/toxicity , Quinolines/toxicity , Rats, Inbred BN , Rats, Long-Evans , Receptors, Neurotensin/antagonists & inhibitors , Retention, Psychology/drug effects , Spatial Memory/drug effects
17.
J Mol Neurosci ; 52(1): 37-47, 2014 Jan.
Article En | MEDLINE | ID: mdl-24395090

VIP is highly expressed in the colon and regulates motility, vasodilatation, and sphincter relaxation. However, its role in the development and progress of colitis is still controversial. Our aim was to determine the participation of VIP on dextran sodium sulfate (DSS)-induced colonic mucosal inflammation using VIP(-/-) and WT mice treated with VIP antagonists. Colitis was induced in 32 adult VIP(-/-) and 14 age-matched WT litter-mates by giving 2.5 % DSS in the drinking water. DSS-treated WT mice were injected daily with VIP antagonists, VIPHyb (n = 22), PG 97-269 (n = 9), or vehicle (n = 31). After euthanasia, colons were examined; colonic cytokines mRNA were quantified. VIP(-/-) mice were remarkably resistant to DSS-induced colitis compared to WT. Similarly, DSS-treated WT mice injected with VIPHyb (1 µM) or PG 97-269 (1 nM) had significantly reduced clinical signs of colitis. Furthermore, colonic expression of IL-1ϐ, TNF-α, and IL-6 was significantly lower in VIP(-/-) and VIPHyb or PG 97-269 compared to vehicle-treated WT. Genetic deletion of VIP or pharmacological inhibition of VIP receptors resulted in resistance to colitis. These data demonstrate a pro-inflammatory role for VIP in murine colitis and suggest that VIP antagonists may be an effective clinical treatment for human inflammatory bowel diseases.


Colitis/drug therapy , Intestinal Mucosa/drug effects , Neurotensin/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Colitis/chemically induced , Colitis/metabolism , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/toxicity , Gene Deletion , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Neurotensin/genetics , Neurotensin/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/therapeutic use
18.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G301-9, 2014 Feb 15.
Article En | MEDLINE | ID: mdl-24356886

Xenin-25 (Xen) is a neurotensin-related peptide secreted by a subset of glucose-dependent insulinotropic polypeptide (GIP)-producing enteroendocrine cells. In animals, Xen regulates gastrointestinal function and glucose homeostasis, typically by initiating neural relays. However, little is known about Xen action in humans. This study determines whether exogenously administered Xen modulates gastric emptying and/or insulin secretion rates (ISRs) following meal ingestion. Fasted subjects with normal (NGT) or impaired (IGT) glucose tolerance and Type 2 diabetes mellitus (T2DM; n = 10-14 per group) ingested a liquid mixed meal plus acetaminophen (ACM; to assess gastric emptying) at time zero. On separate occasions, a primed-constant intravenous infusion of vehicle or Xen at 4 (Lo-Xen) or 12 (Hi-Xen) pmol · kg(-1) · min(-1) was administered from zero until 300 min. Some subjects with NGT received 30- and 90-min Hi-Xen infusions. Plasma ACM, glucose, insulin, C-peptide, glucagon, Xen, GIP, and glucagon-like peptide-1 (GLP-1) levels were measured and ISRs calculated. Areas under the curves were compared for treatment effects. Infusion with Hi-Xen, but not Lo-Xen, similarly delayed gastric emptying and reduced postprandial glucose levels in all groups. Infusions for 90 or 300 min, but not 30 min, were equally effective. Hi-Xen reduced plasma GLP-1, but not GIP, levels without altering the insulin secretory response to glucose. Intense staining for Xen receptors was detected on PGP9.5-positive nerve fibers in the longitudinal muscle of the human stomach. Thus Xen reduces gastric emptying in humans with and without T2DM, probably via a neural relay. Moreover, endogenous GLP-1 may not be a major enhancer of insulin secretion in healthy humans under physiological conditions.


Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Gastric Emptying/drug effects , Hypoglycemic Agents/therapeutic use , Neurotensin/therapeutic use , Postprandial Period , Adult , Biomarkers/blood , Blood Glucose/metabolism , C-Peptide/blood , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/physiopathology , Drug Administration Schedule , Female , Glucagon/blood , Glucagon-Like Peptide 1/blood , Humans , Hypoglycemic Agents/administration & dosage , Infusions, Intravenous , Insulin/blood , Male , Middle Aged , Missouri , Neurotensin/administration & dosage , Receptors, Neurotensin/drug effects , Receptors, Neurotensin/metabolism , Time Factors , Treatment Outcome
19.
Blood ; 121(12): 2347-51, 2013 Mar 21.
Article En | MEDLINE | ID: mdl-23325838

Cytomegalovirus (CMV) infection following allogeneic bone marrow transplant (allo-BMT) is controlled by donor-derived cellular immunity. Vasoactive intestinal peptide (VIP) suppresses Th1 immunity. We hypothesized that blocking VIP-signaling would enhance anti-CMV immunity in murine recipients of allo-BMT. Recipients were transplanted with bone marrow (BM) and T-cells from major histocompatibility complex (MHC)-mismatched VIP-knockout (KO) or wild-type donors, and treated with 7 daily subcutaneous injections of VIPhyb (peptidic VIP-antagonist) or phosphate-buffered saline (PBS). Genetic and pharmacological blockade of VIP-signaling protected allo-BMT recipients from lethal murine CMV (mCMV) infection, improving survival without increasing graft-versus-host disease. Mice treated with VIPhyb or transplanted with VIP-KO allografts had significantly lower viral loads, increased numbers of mCMV-M45-peptide-MHC-tetramer(+) CD8(+) T-cells, with lower PD-1 expression, and enhanced primary and secondary cellular immune responses after mCMV infection than did PBS-treated mice. These results demonstrate that administration of a VIP antagonist after allo-BMT is a promising safely therapeutic approach to enhance antiviral cellular immunity.


Adaptive Immunity/drug effects , Bone Marrow Transplantation/mortality , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Neurotensin/pharmacology , Vasoactive Intestinal Peptide/antagonists & inhibitors , Adaptive Immunity/genetics , Animals , Bone Marrow Transplantation/immunology , Cells, Cultured , Graft vs Host Disease/complications , Graft vs Host Disease/drug therapy , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Herpesviridae Infections/drug therapy , Herpesviridae Infections/genetics , Herpesviridae Infections/mortality , Immunocompromised Host/drug effects , Immunocompromised Host/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/drug effects , Neurotensin/therapeutic use , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects , Survival Analysis , Transplantation, Homologous , Up-Regulation/drug effects , Up-Regulation/immunology , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/pharmacology , Vasoactive Intestinal Peptide/therapeutic use , Viral Load/drug effects
20.
Nat Prod Rep ; 28(4): 741-62, 2011 Apr.
Article En | MEDLINE | ID: mdl-21340067

Anticonvulsant neuropeptides are best known for their ability to suppress seizures and modulate pain pathways. Galanin, neuropeptide Y, somatostatin, neurotensin, dynorphin, among others, have been validated as potential first-in-class anti-epileptic or/and analgesic compounds in animal models of epilepsy and pain, but their therapeutic potential extends to other neurological indications, including neurodegenerative and psychatric disorders. Disease-modifying properties of neuropeptides make them even more attractive templates for developing new-generation neurotherapeutics. Arguably, efforts to transform this class of neuropeptides into drugs have been limited compared to those for other bioactive peptides. Key challenges in developing neuropeptide-based anticonvulsants are: to engineer optimal receptor-subtype selectivity, to improve metabolic stability and to enhance their bioavailability, including penetration across the blood­brain barrier (BBB). Here, we summarize advances toward developing systemically active and CNS-penetrant neuropeptide analogs. Two main objectives of this review are: (1) to provide an overview of structural and pharmacological properties for selected anticonvulsant neuropeptides and their analogs and (2) to encourage broader efforts to convert these endogenous natural products into drug leads for pain, epilepsy and other neurological diseases.


Anticonvulsants , Nervous System Diseases/drug therapy , Neuropeptides , Analgesics, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Blood-Brain Barrier/metabolism , Dynorphins/metabolism , Dynorphins/therapeutic use , Epilepsy/drug therapy , Galanin/metabolism , Galanin/therapeutic use , Molecular Structure , Neuropeptide Y/metabolism , Neuropeptide Y/therapeutic use , Neuropeptides/metabolism , Neuropeptides/therapeutic use , Neurotensin/metabolism , Neurotensin/therapeutic use , Seizures/drug therapy , Sequence Homology, Amino Acid , Somatostatin/metabolism , Somatostatin/therapeutic use
...