Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Sci Rep ; 10(1): 15235, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943676

ABSTRACT

Pharmacological experiments indicate that neuropeptides can effectively tune neuronal activity and modulate locomotor output patterns. However, their functions in shaping innate locomotion often remain elusive. For example, somatostatin has been previously shown to induce locomotion when injected in the brain ventricles but to inhibit fictive locomotion when bath-applied in the spinal cord in vitro. Here, we investigated the role of somatostatin in innate locomotion through a genetic approach by knocking out somatostatin 1.1 (sst1.1) in zebrafish. We automated and carefully analyzed the kinematics of locomotion over a hundred of thousand bouts from hundreds of mutant and control sibling larvae. We found that the deletion of sst1.1 did not impact acousto-vestibular escape responses but led to abnormal exploration. sst1.1 mutant larvae swam over larger distance, at higher speed and performed larger tail bends, indicating that Somatostatin 1.1 inhibits spontaneous locomotion. Altogether our study demonstrates that Somatostatin 1.1 innately contributes to slowing down spontaneous locomotion.


Subject(s)
Somatostatin/physiology , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Biomechanical Phenomena , Exploratory Behavior/physiology , Female , Frameshift Mutation , Gene Knockout Techniques , Larva/physiology , Locomotion/physiology , Male , Sequence Deletion , Somatostatin/deficiency , Somatostatin/genetics , Swimming/physiology , Video Recording , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
2.
Sleep ; 42(10)2019 10 09.
Article in English | MEDLINE | ID: mdl-31328777

ABSTRACT

Slow-wave activity (SWA) is an oscillatory neocortical activity occurring in the electroencephalogram delta (δ) frequency range (~0.5-4 Hz) during nonrapid eye movement sleep. SWA is a reliable indicator of sleep homeostasis after acute sleep loss and is involved in memory processes. Evidence suggests that cortical neuronal nitric oxide synthase (nNOS) expressing neurons that coexpress somatostatin (SST) play a key role in regulating SWA. However, previous studies lacked selectivity in targeting specific types of neurons that coexpress nNOS-cells which are activated in the cortex after sleep loss. We produced a mouse model that knocks out nNOS expression in neurons that coexpress SST throughout the cortex. Mice lacking nNOS expression in SST positive neurons exhibited significant impairments in both homeostatic low-δ frequency range SWA production and a recognition memory task that relies on cortical input. These results highlight that SST+/nNOS+ neurons are involved in the SWA homeostatic response and cortex-dependent recognition memory.


Subject(s)
Cerebral Cortex/metabolism , Delta Rhythm/physiology , Memory/physiology , Nitric Oxide Synthase Type I/deficiency , Recognition, Psychology/physiology , Somatostatin/deficiency , Animals , Electroencephalography/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Nitric Oxide Synthase Type I/genetics , Sleep/physiology , Somatostatin/genetics
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(1): 147-150, 2017 Jan.
Article in Chinese | MEDLINE | ID: mdl-28612576

ABSTRACT

OBJECTIVES: To investigate clinical features in infants of breast milk allergy(BMA), and the possible relationship with the changes of somatostatin (SST) and motilin (MTL) in breast milk. METHODS: Twenty three cases of pure breast feeding infants with allergic gastroenteritis were collected, while another 23 normal infants with pure breast feeding were enrolled as normal controls. Samples of infant stools and breast milk were collected for the measurement of SST and MTL levels detected by by radioimmunity. RESULTS: The levels of SST and MTL in stool samples (pg/mg) were 32.6±8.9, 2.3±3.7 in BMA group and 56.2±12.7, 21.6±4.7 in normal control group, respectively. Those in breast milk (pg/mg) were 236.7±28.9, 159.4±36.7 in BMA group and 412.6±36.7, 216.8±59.7 in normal control group, respectively. All the differences were statistically significant ( P<0.05). In BMA infants, the clinical features were 91.3% (20/23) of diarrhea, 86.9% (21/23) of vomiting, 69.6% (16/23) of hematochezia, 95.7% (22/23) of C-reactive protein (CRP) increasing, 87.0% (20/23) of occult blood in stools, 73.9% (17/23) of neutrophil increasing, 39.1% (9/23) of WBC in stools. CONCLUSIONS: For those infants of breast feeding with persisting and repeated gastrointestinal symptoms, allergy for breast milk should be considered. Deficiency of SST and MTL in breast milk may be a possible cause for food allergy.


Subject(s)
Milk Hypersensitivity , Milk, Human/chemistry , Motilin/analysis , Somatostatin/analysis , Breast Feeding , Female , Humans , Infant , Motilin/deficiency , Somatostatin/deficiency
4.
J Neurovirol ; 22(6): 747-762, 2016 12.
Article in English | MEDLINE | ID: mdl-27178324

ABSTRACT

Memory deficits are characteristic of HIV-associated neurocognitive disorders (HAND) and co-occur with hippocampal pathology. The HIV-1 transactivator of transcription (Tat), a regulatory protein, plays a significant role in these events, but the cellular mechanisms involved are poorly understood. Within the hippocampus, diverse populations of interneurons form complex networks; even subtle disruptions can drastically alter synaptic output, resulting in behavioral dysfunction. We hypothesized that HIV-1 Tat would impair cognitive behavior and injure specific hippocampal interneuron subtypes. Male transgenic mice that inducibly expressed HIV-1 Tat (or non-expressing controls) were assessed for cognitive behavior or had hippocampal CA1 subregions evaluated via interneuron subpopulation markers. Tat exposure decreased spatial memory in a Barnes maze and mnemonic performance in a novel object recognition test. Tat reduced the percentage of neurons expressing neuronal nitric oxide synthase (nNOS) without neuropeptide Y immunoreactivity in the stratum pyramidale and the stratum radiatum, parvalbumin in the stratum pyramidale, and somatostatin in the stratum oriens, which are consistent with reductions in interneuron-specific interneuron type 3 (IS3), bistratified, and oriens-lacunosum-moleculare interneurons, respectively. The findings reveal that an interconnected ensemble of CA1 nNOS-expressing interneurons, the IS3 cells, as well as subpopulations of parvalbumin- and somatostatin-expressing interneurons are preferentially vulnerable to HIV-1 Tat. Importantly, the susceptible interneurons form a microcircuit thought to be involved in feedback inhibition of CA1 pyramidal cells and gating of CA1 pyramidal cell inputs. The identification of vulnerable CA1 hippocampal interneurons may provide novel insight into the basic mechanisms underlying key functional and neurobehavioral deficits associated with HAND.


Subject(s)
CA1 Region, Hippocampal/metabolism , Cognitive Dysfunction/genetics , Interneurons/metabolism , Nitric Oxide Synthase Type I/genetics , Parvalbumins/genetics , Somatostatin/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , CA1 Region, Hippocampal/physiopathology , Cognition/physiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Gene Expression Regulation , Interneurons/pathology , Male , Maze Learning , Mice , Mice, Transgenic , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Nitric Oxide Synthase Type I/deficiency , Parvalbumins/deficiency , Signal Transduction , Somatostatin/deficiency , Transgenes , tat Gene Products, Human Immunodeficiency Virus/metabolism
5.
PLoS One ; 10(3): e0120955, 2015.
Article in English | MEDLINE | ID: mdl-25806796

ABSTRACT

Locally produced growth hormone (GH) and IGF-I are key factors in the regulation of mammary gland (MG) development and may be important in breast cancer development/progression. Somatostatin (SST) and cortistatin (CORT) regulate GH/IGF-I axis at various levels, but their role in regulating GH/IGF-I in MGs remains unknown. Since obesity alters the expression of these systems in different tissues and is associated to MG (patho) physiology, we sought to investigate the role of SST/CORT in regulating GH/IGF-I system in the MGs of lean and obese mice. Therefore, we analyzed GH/IGF-I as well as SST/CORT and ghrelin systems expression in the mammary fat pads (MFPs) of SST- or CORT-knockout (KO) mice and their respective littermate-controls fed a low-fat (LF) or a high-fat (HF) diet for 16 wks. Our results demonstrate that the majority of the components of GH/IGF-I, SST/CORT and ghrelin systems are locally expressed in mouse MFP. Expression of elements of the GH/IGF-I axis was significantly increased in MFPs of HF-fed control mice while lack of endogenous SST partially suppressed, and lack of CORT completely blunted, the up-regulation observed in obese WT-controls. Since SST/CORT are known to exert an inhibitory role on the GH/IGFI axis, the increase in SST/CORT-receptor sst2 expression in MFPs of HF-fed CORT- and SST-KOs together with an elevation on circulating SST in CORT-KOs could explain the differences observed. These results offer new information on the factors (GH/IGF-I axis) involved in the endocrine/metabolic dysregulation of MFPs in obesity, and suggest that CORT is not a mere SST sibling in regulating MG physiology.


Subject(s)
Adipose Tissue/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/metabolism , Neuropeptides/metabolism , Somatostatin/metabolism , Animals , Body Weight , Diet, High-Fat , Female , Ghrelin/metabolism , Leptin/blood , Mice , Mice, Knockout , Mice, Obese , Neuropeptides/deficiency , Neuropeptides/genetics , Obesity/metabolism , Obesity/pathology , Real-Time Polymerase Chain Reaction , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/deficiency , Somatostatin/genetics , Up-Regulation
6.
Horm Metab Res ; 47(1): 56-63, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25350519

ABSTRACT

Early postnatal life is a critical period for development of the endocrine pancreas, involving remodelling of islet cells and maturation of secretory responses. Factors that regulate these processes are undefined. Somatostatin-secreting delta-cells are abundant in the developing pancreas and, because somatostatin inhibits growth, the hormone may regulate islet expansion in early life. The aim of this study was to investigate effects of somatostatin-deficiency on proliferation, apoptosis and pancreas expansion in the first 3 weeks of life in mice. Pancreases from control or somatostatin-knockout mice were analysed for beta cell, alpha cell and pancreatic volumes by morphometry, proliferation by BrdU incorporation and apoptosis by TUNEL labelling. Signalling pathways associated with proliferation and apoptosis were studied by immunohistochemistry and Western blotting. Knockout mice grew normally in the first 3 weeks of life, but had high circulating insulin that normalised by day 21. Beta cell, alpha cell and pancreatic volumes were decreased in knockout mice, accompanied by reduced proliferation and increased apoptosis in the pancreas. Decreased growth was not due to impaired Akt signalling, as Akt phosphorylation and nuclear cyclin-D2 increased in the knockout pancreas. Levels of TGF-ß1, a factor implicated in tissue remodelling, together with SMAD phosphorylation through which TGF-ß mediates its effects, were increased in the knockout pancreas. Beta cell expansion was impaired in knockout mice, potentially compensating for increased insulin secretion from islets lacking inhibitory effects of somatostatin, and was associated with increased TGF-ß1 levels. TGF-ß1 may represent an important regulator of beta cell mass in early life.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Signal Transduction , Somatostatin/deficiency , Transforming Growth Factor beta/metabolism , Animals , Animals, Newborn , Apoptosis , Body Weight , Bromodeoxyuridine/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , In Situ Nick-End Labeling , Male , Mice , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad Proteins/metabolism , Somatostatin/metabolism
7.
Nat Commun ; 5: 5333, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25354876

ABSTRACT

The cellular diversity of interneurons in the neocortex is thought to reflect subtype-specific roles of cortical inhibition. Here we ask whether perturbations to two subtypes--parvalbumin-positive (PV+) and somatostatin-positive (SST+) interneurons--can be compensated for with respect to their contributions to cortical development. We use a genetic cell fate switch to delete both PV+ and SST+ interneurons selectively in cortical layers 2-4 without numerically changing the total interneuron population. This manipulation is compensated for at the level of synaptic currents and receptive fields (RFs) in the somatosensory cortex. By contrast, we identify a deficit in inhibitory synchronization in vitro and a large reduction in cortical gamma oscillations in vivo. This reveals that, while the roles of inhibition in establishing cortical inhibitory/excitatory balance and RFs can be subserved by multiple interneuron subtypes, gamma oscillations depend on cellular properties that cannot be compensated for--likely, the fast signalling properties of PV+ interneurons.


Subject(s)
Cortical Synchronization , Interneurons/physiology , Neocortex/physiology , Pyramidal Cells/physiology , Animals , In Vitro Techniques , Male , Mice , Nuclear Proteins/deficiency , Parvalbumins/deficiency , Patch-Clamp Techniques , Somatostatin/deficiency , Thyroid Nuclear Factor 1 , Transcription Factors/deficiency
8.
Eur J Endocrinol ; 168(4): 491-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23288882

ABSTRACT

OBJECTIVE: Somatotroph adenomas causing acromegaly are histologically classified into densely granulated (DG) and sparsely granulated (SG) subtypes with different morphology, clinical characteristics and treatment outcomes. Granulation pattern has been reported to co-segregate with a recurrent mutation at codon 49 in growth hormone receptor (GHR) and GSP oncogene. This study examines response to the octreotide suppression test (OST) in relation to granulation pattern and mutation in GHR and GSP. DESIGN: This is a retrospective, single-centre study of 52 patients with pathologically confirmed somatotroph adenoma who were naïve to medical therapy presenting between January 2001 and October 2010. METHODS: Clinical, radiological and hormonal data at diagnosis were recorded. GHR and GSP were genotyped, granulation pattern determined and response to the OST measured. RESULTS: SG adenomas were larger (P=0.038), occurred in younger patients (P=0.029), were more common in females (P=0.026) and were more invasive (P<0.0001 and P=0.001), with diminished responses to the OST (P=0.007) compared with DG adenomas. GSP mutation was unrelated to granulation pattern but associated with smaller tumours (P=0.027), producing more GH (P=0.048) that responded better to the OST (P=0.022). Codon 49 of GHR was not mutated. CONCLUSIONS: Adenoma histological phenotype, not genotype, corresponds to clinical and biochemical characteristics and response to the OST. SG adenomas constitute a clinically more unfavourable subtype but are not associated with GHR mutations in our series. Ascertainment of the adenoma subtype may become an important consideration in the management of acromegaly.


Subject(s)
Adenoma/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , Granulation Tissue/pathology , Growth Hormone-Secreting Pituitary Adenoma/pathology , Mutation/genetics , Receptors, Somatotropin , Somatostatin/deficiency , Adenoma/drug therapy , Adenoma/genetics , Adult , Aged , Aged, 80 and over , Chromogranins , Granulation Tissue/physiology , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/genetics , Humans , Middle Aged , Receptors, Somatotropin/genetics , Retrospective Studies , Somatostatin/metabolism , Young Adult
10.
Diabetes ; 58(2): 403-11, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18984743

ABSTRACT

OBJECTIVE: Somatostatin (SST) is secreted by islet delta-cells and by extraislet neuroendocrine cells. SST receptors have been identified on alpha- and beta-cells, and exogenous SST inhibits insulin and glucagon secretion, consistent with a role for SST in regulating alpha- and beta-cell function. However, the specific intraislet function of delta-cell SST remains uncertain. We have used Sst(-/-) mice to investigate the role of delta-cell SST in the regulation of insulin and glucagon secretion in vitro and in vivo. RESEARCH DESIGN AND METHODS: Islet morphology was assessed by histological analysis. Hormone levels were measured by radioimmunoassay in control and Sst(-/-) mice in vivo and from isolated islets in vitro. RESULTS: Islet size and organization did not differ between Sst(-/-) and control islets, nor did islet glucagon or insulin content. Sst(-/-) mice showed enhanced insulin and glucagon secretory responses in vivo. In vitro stimulus-induced insulin and glucagon secretion was enhanced from perifused Sst(-/-) islets compared with control islets and was inhibited by exogenous SST in Sst(-/-) but not control islets. No difference in the switch-off rate of glucose-stimulated insulin secretion was observed between genotypes, but the cholinergic agonist carbamylcholine enhanced glucose-induced insulin secretion to a lesser extent in Sst(-/-) islets compared with controls. Glucose suppressed glucagon secretion from control but not Sst(-/-) islets. CONCLUSIONS: We suggest that delta-cell SST exerts a tonic inhibitory influence on insulin and glucagon secretion, which may facilitate the islet response to cholinergic activation. In addition, delta-cell SST is implicated in the nutrient-induced suppression of glucagon secretion.


Subject(s)
Islets of Langerhans/metabolism , Somatostatin-Secreting Cells/metabolism , Somatostatin/physiology , Animals , Female , Glucagon/metabolism , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Radioimmunoassay , Somatostatin/deficiency , Somatostatin/genetics
11.
Behav Brain Res ; 181(1): 85-95, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17477978

ABSTRACT

This study examined the effect of both separate and combined depletion of brain somatostatin and serotonin on acquisition of the water maze (WM) task. Naïve male Long-Evans rats received injections of p-chlorophenylalanine (PCPA; 500mg/kgx2) to deplete serotonin or cysteamine (90 or 200mg/kg) to deplete somatostatin, or both treatments prior to spatial and reversal training in the water maze. Some rats first received Morris' nonspatial pretraining to train them in the behavioral strategies that are required for successful spatial place learning in this task, prior to drug treatment and spatial training. A detailed behavioral analysis indicated that somatostatin or serotonin depletion alone caused little or no impairment in naïve animals. Depletion of both somatostatin and serotonin in naïve rats impaired performance, with differences in the impairments that depended on the dose of cysteamine. Nonspatially pretrained rats were not impaired. Thus, neither somatostatin nor serotonin alone is crucial for the water maze task, but impairments occur if both somatostatin and serotonin are depleted in naïve rats. The results indicate that some of the performance impairment was due to strategies impairment rather than a spatial place learning impairment. Depletion of both somatostatin and serotonin in naïve rats produces results comparable to the spatial navigation deficits seen in some Alzheimer patients and suggests that combinations of antagonist treatments may better model this disorder than single antagonist treatments do.


Subject(s)
Learning Disabilities/physiopathology , Maze Learning/physiology , Serotonin/deficiency , Somatostatin/deficiency , Animals , Behavior, Animal , Cysteamine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Fenclonine/pharmacology , Learning Disabilities/chemically induced , Male , Maze Learning/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Long-Evans , Scopolamine/pharmacology , Serotonin Antagonists/pharmacology , Time Factors
13.
Clin Endocrinol (Oxf) ; 66(3): 341-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17302866

ABSTRACT

OBJECTIVE: Growth hormone (GH) circulating levels are highly dependent not only on GH secretion rate from the pituitary, but also on the hormone distribution in the compartments of the body and elimination phenomena. In adult GH-deficient patients these factors become critical nowadays, especially when recombinant human GH (rhGH) is available for replacement therapy. In the present study, we assess the influence of both distribution and elimination phenomena on GH pharmacokinetics in adult GH-deficient patients. METHODS: We used a four-step methodology following a compartmental approach after an intravenous bolus of recombinant GH in adult GH-deficient patients. RESULTS: We found that GH kinetics are clearly explained by a bi-exponential, two-compartmental model in GH-deficient patients, similarly than in normal or diabetic subjects, as previously shown. We have also observed a marked delay in the whole GH elimination process in GH-deficient patients compared to normal adult subjects, as revealed by metabolic clearance ratio (MCR), elimination constant from central compartment (k(10)), and mean resident time in the body (MRT). Interestingly, such a delay appear to be caused by deep changes in the distribution phase (Mtt(1)- mean transit time-1; T(1/2alpha)- GH half-life at distribution phase), while the elimination phenomenon remains unaltered. CONCLUSION: Our results emphasize the relevance of distribution phenomena in GH pharmacokinetics, and indicates that studies avoiding data from the GH distribution phase, such as those carried out in steady-state conditions, or those using noncompartmental models, could easily miss relevant information. Our data should be taken into consideration when establishing the appropriate dosage for GH replacement treatments in GH-deficient patients, and calculations should include GH distribution kinetics.


Subject(s)
Hormone Replacement Therapy/methods , Human Growth Hormone/pharmacokinetics , Somatostatin/deficiency , Adult , Aged , Case-Control Studies , Drug Administration Schedule , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Half-Life , Human Growth Hormone/therapeutic use , Humans , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Models, Statistical , Octreotide/pharmacology
14.
Clin Endocrinol (Oxf) ; 65(6): 783-91, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17121531

ABSTRACT

OBJECTIVE: The aim of the study was to clarify whether a gender difference exists with respect to bone mineral density (BMD) and bone mineral content (BMC) in adult patients with growth hormone deficiency (GHD). DESIGN: A case-control design. METHODS: Blood sampling for measurements of calcium, phosphate, creatinine, PTH, vitamin D, IGF-1, markers of bone formation and bone resorption, and dual energy X-ray absorptiometry (DEXA), to determine BMD and BMC of the lumbar spine, hip, distal arm and total body, were performed in 34 patients with GHD (19 females) and 34 sex-, age- and weight-matched healthy control subjects. The patients were well substituted on all pituitary axes, apart from GH. RESULTS: GH-deficient males had significantly lower BMD in the lumbar spine (P = 0.02), hip (P = 0.01) and total body (P = 0.003) than healthy males while GH-deficient females compared to healthy females had identical BMD values at all regions. This gender difference was even more obvious when BMD values were expressed as Z-scores or as three-dimensional BMD of the total body. The bone formation and bone resorption markers, as well as calcium and vitamin D, were all at the same levels in GH-deficient and healthy males, indicating identical bone turnover. The GH-deficient females, however, had significantly lower levels of bone markers compared to healthy females, indicating a reduced bone turnover. Oestrogen substitution of the GH-deficient females could explain this difference. CONCLUSIONS: Compared to healthy control subjects GH-deficient males had, in contrast to GH-deficient females, significantly reduced BMD and BMC. This obvious gender difference seems to be caused by the oestrogen substitution given to the females, compensating for the lack of GH, an effect testosterone does not seem to possess.


Subject(s)
Bone Density , Sex Factors , Somatostatin/deficiency , Absorptiometry, Photon , Adult , Aged , Alkaline Phosphatase/blood , Biomarkers/blood , Bone Remodeling , Calcitriol/blood , Case-Control Studies , Collagen Type I/blood , Estradiol/blood , Female , Humans , Insulin-Like Growth Factor I/analysis , Male , Middle Aged , Osteocalcin/blood , Parathyroid Hormone/blood , Peptide Fragments/blood , Peptides/blood , Procollagen/blood , Somatostatin/blood , Statistics, Nonparametric
15.
J Alzheimers Dis ; 9(3 Suppl): 117-9, 2006.
Article in English | MEDLINE | ID: mdl-17004364

ABSTRACT

This brief paper reviews the work on dementia by the Neuropathology group at the Einstein College of Medicine and later at the University of California, San Diego, from the time of our first approaches to Alzheimer Disease in 1959. The electron microscope studies concerned the tangle (got it wrong) and then the plaque (got it right). Lysosomes and active mitochondria were noted in the plaques. Axoplasmic transport was suggested to be abnormal. We studied the plaques in old dogs and old monkeys, and then went on to use image analysis to count neurons in the neocortex of Alzheimer cases and in examples of normal aging. Later in San Diego we quantified presynaptic boutons and recognized their loss as the major direct cause of dementia. Many collaborators including Henry Wisniewski participated in these early attempts to understand the disease.


Subject(s)
Alzheimer Disease/history , Alzheimer Disease/pathology , Brain/pathology , Research/history , Aging/physiology , Alzheimer Disease/physiopathology , Animals , Cell Count , Cell Death/physiology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , History, 20th Century , Humans , Receptors, Presynaptic/physiology , Somatostatin/deficiency , United States
16.
Eur J Neurosci ; 23(9): 2404-22, 2006 May.
Article in English | MEDLINE | ID: mdl-16706848

ABSTRACT

Somatostatin-14 (SRIF) co-localizes with gamma-aminobutyric acid (GABA) in the hippocampus and regulates neuronal excitability. A role of SRIF in the control of seizures has been proposed, although its exact contribution requires some clarification. In particular, SRIF knockout (KO) mice do not exhibit spontaneous seizures, indicating that compensatory changes may occur in KO. In the KO hippocampus, we examined whether specific SRIF receptors and/or the cognate peptide cortistatin-14 (CST) compensate for the absence of SRIF. We found increased levels of both sst2 receptors (sst2) and CST, and we explored the functional consequences of sst2 compensation on bursting activity and synaptic responses in hippocampal slices. Bursting was decreased by SRIF in wild-type (WT) mice, but it was not affected by either CST or sst2 agonist and antagonist. sst4 agonist increased bursting frequency in either WT or KO. In WT, but not in KO, its effects were blocked by agonizing or antagonizing sst2, suggesting that sst2 and sst4 are functionally coupled in the WT hippocampus. Bursting was reduced in KO as compared with WT and was increased upon application of sst2 antagonist, while SRIF, CST and sst2 agonist had no effect. At the synaptic level, we observed that in WT, SRIF decreased excitatory postsynaptic potentials which were, in contrast, increased by sst2 antagonist in KO. We conclude that sst2 compensates for SRIF absence and that its upregulation is responsible for reduced bursting and decreased excitatory transmission in KO mice. We suggest that a critical density of sst2 is needed to control hippocampal activity.


Subject(s)
Action Potentials/genetics , Hippocampus/physiology , Receptors, Somatostatin/physiology , Somatostatin/deficiency , Synaptic Transmission/genetics , Up-Regulation/genetics , Adenylyl Cyclases/metabolism , Animals , Blotting, Western/methods , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Binding/drug effects , Protein Binding/physiology , RNA, Messenger/metabolism , Radioligand Assay/methods , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction/methods , Somatostatin/analogs & derivatives , Synaptic Transmission/drug effects , Up-Regulation/drug effects
17.
Neuroreport ; 15(12): 1979-82, 2004 Aug 26.
Article in English | MEDLINE | ID: mdl-15305149

ABSTRACT

The effect of Gly-Pro-Glu (GPE) on the somatostatinergic system of the temporal cortex in amyloid beta-peptide (Abeta) treated rats was investigated. Intracerebroventricular Abeta25-35 administration for 14 days (300 pmol/day) to ovariectomized rats produced a marked reduction in somatostatin (SRIF) content, SRIF receptor density and reduced the inhibitory effect of SRIF on adenylyl cyclase activity. I.p. injection of three doses (300 microg) of GPE on days 0, 6 and 12 resulted in a partial recovery of the parameters affected by Abeta25-35 administration. These results indicate that GPE may have an in vivo effect protecting the temporal cortical somatostatinergic system from Abeta insult.


Subject(s)
Amyloid beta-Peptides/pharmacology , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Somatostatin/drug effects , Temporal Lobe/drug effects , Temporal Lobe/metabolism , Adenylyl Cyclases/metabolism , Amyloid beta-Peptides/administration & dosage , Analysis of Variance , Animals , Binding Sites , Blotting, Western/methods , Dose-Response Relationship, Drug , Drug Interactions , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Ovariectomy/methods , Peptide Fragments/administration & dosage , Rats , Rats, Wistar , Receptors, Somatostatin/metabolism , Somatostatin/deficiency , Time Factors
18.
Neuroscience ; 122(2): 437-47, 2003.
Article in English | MEDLINE | ID: mdl-14614908

ABSTRACT

A somatostatin deficit occurs in the cerebral cortex of Alzheimer's disease patients without a major loss in somatostatin-containing neurons. This deficit could be related to a reduction in the rate of proteolytic processing of peptide precursors. Since the two proprotein convertases (PC)1 and PC2 are responsible for the processing of neuropeptide precursors directed to the regulated secretory pathway, we examined whether they are involved first in the proteolytic processing of prosomatostatin in mouse and human brain and secondly in somatostatin defect associated with Alzheimer's disease. By size exclusion chromatography, the cleavage of prosomatostatin to somatostatin-14 is almost totally abolished in the cortex of PC2 null mice, while the proportions of prosomatostatin and somatostatin-28 are increased. By immunohistochemistry, PC1 and PC2 were localized in many neuronal elements in human frontal and temporal cortex. The convertases levels were quantified by Western blot, as well as the protein 7B2 which is required for the production of active PC2. No significant change in PC1 levels was observed in Alzheimer's disease. In contrast, a marked decrease in the ratio of the PC2 precursor to the total enzymatic pool was observed in the frontal cortex of Alzheimer patients. This decrease coincides with an increase in the binding protein 7B2. However, the content and enzymatic activity of the PC2 mature form were similar in Alzheimer patients and controls. Therefore, the cortical somatostatin defect is not due to convertase alteration occuring during Alzheimer's disease. Further studies will be needed to assess the mechanisms involved in somatostatin deficiency in Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Proprotein Convertase 2/physiology , Protein Precursors/metabolism , Protein Processing, Post-Translational/physiology , Somatostatin/biosynthesis , Somatostatin/deficiency , Somatostatin/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Analysis of Variance , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Female , Humans , Linear Models , Male , Mice , Mice, Knockout , Proprotein Convertase 2/deficiency , Proprotein Convertase 2/genetics , Protein Precursors/genetics , Protein Processing, Post-Translational/genetics , Rats , Rats, Sprague-Dawley , Somatostatin/genetics
19.
Proc Natl Acad Sci U S A ; 100(22): 12944-9, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14555768

ABSTRACT

Fifty percent of the world's population is infected with Helicobacter pylori; however, treatment has been insufficient to eradicate the organisms due to rising antibiotic resistance. Helicobacter infection is characterized by induction of a T helper 1 lymphocyte (Th1) immune response, hypergastrinemia, and suppressed tissue somatostatin (SOM) levels. However, the mechanism by which the immune response regulates acid secretion is not known. We show here that treatment with IFN-gamma, a Th1 cytokine, was sufficient to induce gastritis, increase gastrin, and decrease SOM levels within 7 days. In contrast, the T helper 2 lymphocyte cytokine IL-4 increased SOM levels and effectively suppressed gastrin expression and secretion. This result demonstrated reciprocal regulation of acid regulatory peptides by immune modulators. IL-4 pretreatment prevented gastritis in infected wild-type but not in SOM null mice. Thus, the ability of IL-4 to oppose a Th1-mediated infection required SOM. Immunofluorescence was used to document the presence of IL-4 receptors on the gastric SOM-secreting cell (D cell). Moreover, IL-4 stimulated SOM release from primary D cell cultures. Treatment of mice chronically infected with Helicobacter felis for 2 mo with the SOM analogue octreotide resolved the inflammation. Thus, a mechanism by which IL-4 resolves inflammation in the stomach is by stimulating the release of SOM from gastric D cells.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/drug therapy , Helicobacter felis , Interleukin-4/therapeutic use , Somatostatin/physiology , Animals , Gastric Acid , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/drug therapy , Gastritis/immunology , Gastritis/pathology , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Inflammation , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Somatostatin/deficiency , Somatostatin/genetics , Somatostatin/metabolism
20.
Naunyn Schmiedebergs Arch Pharmacol ; 367(6): 562-71, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12759718

ABSTRACT

Five somatostatin receptors (sst) have been cloned and mRNAs for the first four (sst1-4) are expressed in many brain regions. In the present work, we compared the distribution of the non-selective ligand [125I]-Tyr0-DTrp8-SRIF14 by autoradiography in 24 brain regions and pituitary in wild type, sst1- to sst4- or SRIF-gene invalidated (KO) mice. [125I]-Tyr0-DTrp8-SRIF14 binding was not significantly modified in sst1 KO mouse brain with the noticeable exception of the substantia nigra and only moderately decreased in pituitary. For sst2 KO mice, a general decrease (>75%) was observed in most regions, with the noticeable exception of the olfactory bulb and CA1 field of the hippocampus. SST3 KO brain displayed a decrease in binding in the external plexiform layer of the olfactory bulb only (-54%). For sst4 KO mice, [125I]-Tyr0-DTrp8-SRIF14 binding levels in the external plexiform (-35%) and glomerular (-39%) layers of the olfactory bulb as well as the hippocampus CA1 field (-68%) were significantly decreased. In SRIF KO mice, a significant increase in binding levels was observed in olfactory bulb, anterior olfactory nucleus, frontal cortex upper layers, lateral septum, CA1 field, zona incerta and lateral hypothalamus, substantia nigra, periaqueductal grey and parabrachial nucleus. Competition with selective ligands (CH275, octreotide or L-779,976, L-796,778, L-803,087, and octreotide or L-817,778, for sst1-5 receptors, respectively) was in accordance with these findings. Moreover, octreotide was still able to compete on residual [125I]-Tyr0-DTrp8-SRIF14 binding sites in sst2 KO pituitary. It is concluded that most [125I]-Tyr0-DTrp8-SRIF14 binding sites in mouse brain and pituitary belong to the sst2 subtype but for the olfactory bulb (sst3 and sst4 receptors), the CA1 of the hippocampus (sst4 receptors) and the pituitary (sst5 and sst1 receptors) in which other subtypes are also expressed. The overall increase in [125I]-Tyr0-DTrp8-SRIF14 binding in SRIF KO mice indicates that SRIF receptors, mostly from the sst2 subtype, are regulated by the endogenous ligand(s).


Subject(s)
Brain/metabolism , Gene Deletion , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/deficiency , Somatostatin/metabolism , Animals , Female , Iodine Radioisotopes/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Octreotide/metabolism , Protein Binding/genetics , Receptors, Somatostatin/deficiency , Somatostatin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...