1
|
Leibowitz JA, Natarajan G, Zhou J, Carney PR, Ormerod BK. Sustained somatostatin gene expression reverses kindling-induced increases in the number of dividing Type-1 neural stem cells in the hippocampi of behaviorally responsive rats. Epilepsy Res 2019; 150:78-94. [PMID: 30735971 DOI: 10.1016/j.eplepsyres.2019.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/18/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022]
Abstract
Neurogenesis persists throughout life in the hippocampi of all mammals, including humans. In the healthy hippocampus, relatively quiescent Type-1 neural stem cells (NSCs) can give rise to more proliferative Type-2a neural progenitor cells (NPCs), which generate neuronal-committed Type-2b NPCs that mature into Type-3 neuroblasts. Many Type-3 neuroblasts survive and mature into functionally integrated granule neurons over several weeks. In kindling models of epilepsy, neurogenesis is drastically upregulated and many new neurons form aberrant connections that could support epileptogenesis and/or seizures. We have shown that sustained vector-mediated hippocampal somatostatin (SST) expression can both block epileptogenesis and reverse seizure susceptibility in fully kindled rats. Here we test whether adeno-associated virus (AAV) vector-mediated sustained SST expression modulates hippocampal neurogenesis and microglial activation in fully kindled rats. We found significantly more dividing Type-1 NSCs and a corresponding increased number of surviving new neurons in the hippocampi of kindled versus sham-kindled rats. Increased numbers of activated microglia were found in the granule cell layer and hilus of kindled rats at both time points. After intrahippocampal injection with either eGFP or SST-eGFP vector, we found similar numbers of dividing Type-1 NSCs and -2 NPCs and surviving BrdU+ neurons and glia in the hippocampi of kindled rats. Upon observed variability in responses to SST-eGFP (2/4 rats exhibited Grade 0 seizures in the test session), we conducted an additional experiment. We found significantly fewer dividing Type-1 NSCs in the hippocampi of SST-eGFP vector-treated responder rats (5/13 rats) relative to SST-eGFP vector-treated non-responders and eGFP vector-treated controls that exhibited high-grade seizures on the test session. The number of activated microglia was upregulated in the GCL and hilus of kindled rats, regardless of vector treatment. These data support the hypothesis that sustained SST expression exerts antiepileptic effects potentially through normalization of neurogenesis and suggests that abnormally high proliferating Type-1 NSC numbers may be a cellular mechanism of epilepsy.
Collapse
Affiliation(s)
| | - Gowri Natarajan
- Department of Neurology and Pediatrics, USA; Neuroscience Program, USA
| | - Junli Zhou
- Department of Neurology and Pediatrics, USA; Neuroscience Program, USA
| | - Paul R Carney
- Department of Neurology and Pediatrics, USA; Neuroscience Program, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brandi K Ormerod
- J. Crayton Pruitt Family Department of Biomedical Engineering, USA; Department of Neuroscience, USA; McKnight Brain Institute, USA.
| |
Collapse
|
2
|
Lau A, Bourkas M, Lu YQQ, Ostrowski LA, Weber-Adrian D, Figueiredo C, Arshad H, Shoaei SZS, Morrone CD, Matan-Lithwick S, Abraham KJ, Wang H, Schmitt-Ulms G. Functional Amyloids and their Possible Influence on Alzheimer Disease. Discoveries (Craiova) 2017; 5:e79. [PMID: 32309597 PMCID: PMC7159844 DOI: 10.15190/d.2017.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Amyloids play critical roles in human diseases but have increasingly been recognized to also exist naturally. Shared physicochemical characteristics of amyloids and of their smaller oligomeric building blocks offer the prospect of molecular interactions and crosstalk amongst these assemblies, including the propensity to mutually influence aggregation. A case in point might be the recent discovery of an interaction between the amyloid β peptide (Aβ) and somatostatin (SST). Whereas Aβ is best known for its role in Alzheimer disease (AD) as the main constituent of amyloid plaques, SST is intermittently stored in amyloid-form in dense core granules before its regulated release into the synaptic cleft. This review was written to introduce to readers a large body of literature that surrounds these two peptides. After introducing general concepts and recent progress related to our understanding of amyloids and their aggregation, the review focuses separately on the biogenesis and interactions of Aβ and SST, before attempting to assess the likelihood of encounters of the two peptides in the brain, and summarizing key observations linking SST to the pathobiology of AD. While the review focuses on Aβ and SST, it is to be anticipated that crosstalk amongst functional and disease-associated amyloids will emerge as a general theme with much broader significance in the etiology of dementias and other amyloidosis.
Collapse
Affiliation(s)
- Angus Lau
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Matthew Bourkas
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Yang Qing Qin Lu
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Lauren Anne Ostrowski
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Danielle Weber-Adrian
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Carlyn Figueiredo
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Hamza Arshad
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Seyedeh Zahra Shams Shoaei
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Christopher Daniel Morrone
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Stuart Matan-Lithwick
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Karan Joshua Abraham
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Hansen Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Gerold Schmitt-Ulms
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| |
Collapse
|
3
|
Ji L, Wu HT, Qin XY, Lan R. Dissecting carboxypeptidase E: properties, functions and pathophysiological roles in disease. Endocr Connect 2017; 6:R18-R38. [PMID: 28348001 PMCID: PMC5434747 DOI: 10.1530/ec-17-0020] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 01/02/2023]
Abstract
Since discovery in 1982, carboxypeptidase E (CPE) has been shown to be involved in the biosynthesis of a wide range of neuropeptides and peptide hormones in endocrine tissues, and in the nervous system. This protein is produced from pro-CPE and exists in soluble and membrane forms. Membrane CPE mediates the targeting of prohormones to the regulated secretory pathway, while soluble CPE acts as an exopeptidase and cleaves C-terminal basic residues from peptide intermediates to generate bioactive peptides. CPE also participates in protein internalization, vesicle transport and regulation of signaling pathways. Therefore, in two types of CPE mutant mice, Cpefat/Cpefat and Cpe knockout, loss of normal CPE leads to a lot of disorders, including diabetes, hyperproinsulinemia, low bone mineral density and deficits in learning and memory. In addition, the potential roles of CPE and ΔN-CPE, an N-terminal truncated form, in tumorigenesis and diagnosis were also addressed. Herein, we focus on dissecting the pathophysiological roles of CPE in the endocrine and nervous systems, and related diseases.
Collapse
Affiliation(s)
- Lin Ji
- Department of Cell Biology & Medical GeneticsSchool of Medicine, Shenzhen University, Shenzhen, China
| | - Huan-Tong Wu
- Beijing Engineering Research Center of Food Environment and HealthCollege of Life & Environmental Sciences, Minzu University of China, Beijing, China
| | - Xiao-Yan Qin
- Beijing Engineering Research Center of Food Environment and HealthCollege of Life & Environmental Sciences, Minzu University of China, Beijing, China
| | - Rongfeng Lan
- Department of Cell Biology & Medical GeneticsSchool of Medicine, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Natarajan G, Leibowitz JA, Zhou J, Zhao Y, McElroy JA, King MA, Ormerod BK, Carney PR. Adeno-associated viral vector-mediated preprosomatostatin expression suppresses induced seizures in kindled rats. Epilepsy Res 2017; 130:81-92. [PMID: 28167431 DOI: 10.1016/j.eplepsyres.2017.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/04/2016] [Accepted: 01/04/2017] [Indexed: 01/29/2023]
Abstract
Somatostatin is expressed widely in the hippocampus and notably in hilar GABAergic neurons that are vulnerable to seizure neuropathology in chronic temporal lobe epilepsy. We previously demonstrated that sustained bilateral preprosomatostatin (preproSST) expression in the hippocampus prevents the development of generalized seizures in the amygdala kindling model of temporal lobe epilepsy. Here we tested whether sustained preproSST expression is anticonvulsant in rats already kindled to high-grade seizures. Rats were kindled until they exhibited 3 consecutive Racine Grade 5 seizures before adeno-associated virus serotype 5 (AAV5) vector driving either eGFP (AAV5-CBa-eGFP) or preproSST and eGFP (AAV5-CBa-preproSST-eGFP) expression was injected bilaterally into the hippocampal dentate gyrus and CA1 region. Retested 3 weeks later, rats that received control vector (AAV5-CBa-eGFP) continued to exhibit high-grade seizures whereas 6/13 rats that received preproSST vector (AAV5-CBa-preproSST-eGFP) were seizure-free. Of these rats, 5/6 remained seizure-free after repeated stimulation sessions and when the stimulation current was increased. These results suggest that vector-mediated expression of preproSST may be a viable therapeutic strategy for temporal lobe epilepsy.
Collapse
Affiliation(s)
- Gowri Natarajan
- Wilder Center of Excellence for Epilepsy Research, University of Florida, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA; Department of Neurology, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jeffrey A Leibowitz
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Junli Zhou
- Wilder Center of Excellence for Epilepsy Research, University of Florida, Gainesville, FL 32611, USA; Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA; Department of Neurology, University of Florida, Gainesville, FL 32611, USA
| | - Yang Zhao
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Jessica A McElroy
- Wilder Center of Excellence for Epilepsy Research, University of Florida, Gainesville, FL 32611, USA; Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Michael A King
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA; NF/SG VA Medical Center, University of Florida, Gainesville, FL 32611, USA
| | - Brandi K Ormerod
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Paul R Carney
- Wilder Center of Excellence for Epilepsy Research, University of Florida, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA; Department of Neurology, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
5
|
Stijnen P, Ramos-Molina B, O'Rahilly S, Creemers JWM. PCSK1 Mutations and Human Endocrinopathies: From Obesity to Gastrointestinal Disorders. Endocr Rev 2016; 37:347-71. [PMID: 27187081 DOI: 10.1210/er.2015-1117] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prohormone convertase 1/3, encoded by the PCSK1 gene, is a serine endoprotease that is involved in the processing of a variety of proneuropeptides and prohormones. Humans who are homozygous or compound heterozygous for loss-of-function mutations in PCSK1 exhibit a variable and pleiotropic syndrome consisting of some or all of the following: obesity, malabsorptive diarrhea, hypogonadotropic hypogonadism, altered thyroid and adrenal function, and impaired regulation of plasma glucose levels in association with elevated circulating proinsulin-to-insulin ratio. Recently, more common variants in the PCSK1 gene have been found to be associated with alterations in body mass index, increased circulating proinsulin levels, and defects in glucose homeostasis. This review provides an overview of the endocrinopathies and other disorders observed in prohormone convertase 1/3-deficient patients, discusses the possible biochemical basis for these manifestations of the disease, and proposes a model whereby certain missense mutations in PCSK1 may result in proteins with a dominant negative action.
Collapse
Affiliation(s)
- Pieter Stijnen
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Bruno Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephen O'Rahilly
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - John W M Creemers
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
6
|
Steinmetz AB, Johnson SA, Iannitelli DE, Pollonini G, Alberini CM. Insulin-like growth factor 2 rescues aging-related memory loss in rats. Neurobiol Aging 2016; 44:9-21. [PMID: 27318130 DOI: 10.1016/j.neurobiolaging.2016.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/22/2016] [Accepted: 04/12/2016] [Indexed: 10/21/2022]
Abstract
Aging is accompanied by declines in memory performance, and particularly affects memories that rely on hippocampal-cortical systems, such as episodic and explicit. With aged populations significantly increasing, the need for preventing or rescuing memory deficits is pressing. However, effective treatments are lacking. Here, we show that the level of the mature form of insulin-like growth factor 2 (IGF-2), a peptide regulated in the hippocampus by learning, required for memory consolidation and a promoter of memory enhancement in young adult rodents, is significantly reduced in hippocampal synapses of aged rats. By contrast, the hippocampal level of the immature form proIGF-2 is increased, suggesting an aging-related deficit in IGF-2 processing. In agreement, aged compared to young adult rats are deficient in the activity of proprotein convertase 2, an enzyme that likely mediates IGF-2 posttranslational processing. Hippocampal administration of the recombinant, mature form of IGF-2 rescues hippocampal-dependent memory deficits and working memory impairment in aged rats. Thus, IGF-2 may represent a novel therapeutic avenue for preventing or reversing aging-related cognitive impairments.
Collapse
Affiliation(s)
- Adam B Steinmetz
- Center for Neural Science, New York University, New York, NY, USA
| | - Sarah A Johnson
- Center for Neural Science, New York University, New York, NY, USA
| | | | | | | |
Collapse
|
7
|
CPE overexpression is correlated with pelvic lymph node metastasis and poor prognosis in patients with early-stage cervical cancer. Arch Gynecol Obstet 2015; 294:333-42. [PMID: 26695643 DOI: 10.1007/s00404-015-3985-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/07/2015] [Indexed: 12/16/2022]
Abstract
PURPOSE Elevated carboxypeptidase E (CPE) levels play crucial roles in tumorigenesis and metastasis. This study investigated the expression and clinicopathological significance of CPE in early-stage cervical cancer. METHODS Elevated carboxypeptidase E expression was analyzed using quantitative polymerase chain reaction and western blotting in normal cervical tissue, cervical cancer cell lines, and in cervical cancer tissues and adjacent noncancerous tissues (ANTs) from the same patient. Immunohistochemistry (IHC) was used to examine CPE expression in tissue samples from 112 patients with early-stage cervical cancer (FIGO stages Ia2-IIa2), 60 patients with cervical intraepithelial neoplasia, and 19 patients with normal cervical tissues (NCTs). Associations between CPE expression and prognostic and diagnostic factors were evaluated statistically. RESULTS CPE expression was significantly higher in cervical cancer cell lines and tissues than in normal tissues and ANTs. Semi-quantitative analysis of IHC indicated that CPE gradually increased from CIN I to cervical cancer, but was absent in NCTs. CPE expression was seen in 40.2 % (45/112) of the cervical cancer samples. CPE expression was significantly associated with FIGO stage (P = 0.003), tumor size (P = 0.012), stromal invasion (P < 0.001), lymphovascular space invasion (P = 0.016), parametrial infiltration (P = 0.027), vaginal involvement (P = 0.007), postoperative adjuvant therapy (P = 0.024), recurrence (P < 0.001), survival (P < 0.001), and pelvic lymph node metastasis (PLNM) (P < 0.001), and it was significantly higher in tissues from patients with PLNM than without PLNM. Logistic regression analysis identified high-level CPE expression as an independent risk factor for PLNM (P = 0.001). Patients with higher CPE expression had shorter overall survival duration than patients with lower CPE expression. Univariate and multivariate Cox-regression analyses suggested that high-level CPE expression is an independent prognostic factor for overall survival in early-stage cervical cancer. CONCLUSIONS High-level CPE expression was associated with a poor prognosis in early-stage cervical cancer. CPE may serve as a biomarker for predicting PLNM and survival in these patients.
Collapse
|
8
|
Qu CL, Dang YH, Tang JS. Administration of somatostatin analog octreotide in the ventrolateral orbital cortex produces sex-related antinociceptive effects on acute and formalin-induced nociceptive behavior in rats. Neurochem Int 2015; 87:77-84. [PMID: 26055971 DOI: 10.1016/j.neuint.2015.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/10/2015] [Accepted: 06/02/2015] [Indexed: 12/23/2022]
Abstract
The present study was designed to examine whether somatostatin analog octreotide (OCT) was involved in antinociception in the ventrolateral orbital cortex (VLO) and determine whether this effect had a sex difference between male and female rats. The radiant heat-evoked tail flick (TF) reflex was used as an index of acute nociceptive response in lightly anesthetized rats. The number of flinches evoked by formalin injection into the hindpaw was used to evaluate inflammatory persistent pain in conscious rats. Administration of OCT (2.0, 5.0 10.0 ng in 0.5 µl) into the VLO depressed the TF reflex in a dose-dependent manner only in female rats, but not male rats. Pretreatment with a nonselective somatostatin receptor antagonist cyclo-somatostatin (c-SOM) (25.0 µg in 0.5 µl) into the VLO antagonized 10.0 ng OCT-induced inhibition of the TF reflex in female rats. Similarly, application of high dose of OCT (10.0 ng in 0.5 µl) into the VLO depressed formalin-induced flinching response in the early and late phases only in female rats, and had no any effects in male rats. Pretreatment with c-SOM (25.0 µg in 0.5 µl) into the VLO totally antagonized the 10 ng OCT-induced inhibition of the flinches in both phases in female rats. Additionally, single administration of c-SOM into the VLO failed to alter tail reflex latencies and formalin-induced nociceptive behaviors in female rats. The results provide the first valuable evidence that somatostatin and its receptors are involved in antinociception in acute heat-evoked nociception and inflammatory persistent pain only in female rats, not male rats, in the VLO.
Collapse
Affiliation(s)
- Chao-Ling Qu
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China.
| | - Yong-Hui Dang
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Yanta Road West 76#, Xi'an, Shaanxi 710061, China
| | - Jing-Shi Tang
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| |
Collapse
|
9
|
Abstract
Neuropeptides are found in many mammalian CNS neurons where they play key roles in modulating neuronal activity. In contrast to amino acid transmitter release at the synapse, neuropeptide release is not restricted to the synaptic specialization, and after release, a neuropeptide may diffuse some distance to exert its action through a G protein-coupled receptor. Some neuropeptides such as hypocretin/orexin are synthesized only in single regions of the brain, and the neurons releasing these peptides probably have similar functional roles. Other peptides such as neuropeptide Y (NPY) are synthesized throughout the brain, and neurons that synthesize the peptide in one region have no anatomical or functional connection with NPY neurons in other brain regions. Here, I review converging data revealing a complex interaction between slow-acting neuromodulator peptides and fast-acting amino acid transmitters in the control of energy homeostasis, drug addiction, mood and motivation, sleep-wake states, and neuroendocrine regulation.
Collapse
|
10
|
Machida M, Fujimaki S, Hidaka R, Asashima M, Kuwabara T. The insulin regulatory network in adult hippocampus and pancreatic endocrine system. Stem Cells Int 2012; 2012:959737. [PMID: 22988465 PMCID: PMC3440949 DOI: 10.1155/2012/959737] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/06/2012] [Accepted: 08/09/2012] [Indexed: 12/19/2022] Open
Abstract
There is a very strong correlation between the insulin-mediated regulatory system of the central nervous system and the pancreatic endocrine system. There are many examples of the same transcriptional factors being expressed in both regions in their embryonic development stages. Hormonal signals from the pancreatic islets influence the regulation of energy homeostasis by the brain, and the brain in turn influences the secretions of the islets. Diabetes induces neuronal death in different regions of the brain especially hippocampus, causes alterations on the neuronal circuits and therefore impairs learning and memory, for which the hippocampus is responsible. The hippocampus is a region of the brain where steady neurogenesis continues throughout life. Adult neurogenesis from undifferentiated neural stem cells is greatly decreased in diabetic patients, and as a result their learning and memory functions decline. Might it be possible to reactivate stem cells whose functions have deteriorated and that are present in the tissues in which the lesions occur in diabetes, a lifestyle disease, which plagues modern humans and develops as a result of the behavior of insulin-related factor? In this paper we summarize research in regard to these matters based on examples in recent years.
Collapse
Affiliation(s)
| | | | | | | | - Tomoko Kuwabara
- Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-4 Higashi, Tsukuba Science City 305-8562, Japan
| |
Collapse
|
11
|
Cawley NX, Wetsel WC, Murthy SRK, Park JJ, Pacak K, Loh YP. New roles of carboxypeptidase E in endocrine and neural function and cancer. Endocr Rev 2012; 33:216-53. [PMID: 22402194 PMCID: PMC3365851 DOI: 10.1210/er.2011-1039] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/18/2012] [Indexed: 01/14/2023]
Abstract
Carboxypeptidase E (CPE) or carboxypeptidase H was first discovered in 1982 as an enkephalin-convertase that cleaved a C-terminal basic residue from enkephalin precursors to generate enkephalin. Since then, CPE has been shown to be a multifunctional protein that subserves many essential nonenzymatic roles in the endocrine and nervous systems. Here, we review the phylogeny, structure, and function of CPE in hormone and neuropeptide sorting and vesicle transport for secretion, alternative splicing of the CPE transcript, and single nucleotide polymorphisms in humans. With this and the analysis of mutant and knockout mice, the data collectively support important roles for CPE in the modulation of metabolic and glucose homeostasis, bone remodeling, obesity, fertility, neuroprotection, stress, sexual behavior, mood and emotional responses, learning, and memory. Recently, a splice variant form of CPE has been found to be an inducer of tumor growth and metastasis and a prognostic biomarker for metastasis in endocrine and nonendocrine tumors.
Collapse
Affiliation(s)
- Niamh X Cawley
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
12
|
|
13
|
Rajput PS, Kharmate G, Norman M, Liu SH, Sastry BR, Brunicardi CF, Kumar U. Somatostatin receptor 1 and 5 double knockout mice mimic neurochemical changes of Huntington's disease transgenic mice. PLoS One 2011; 6:e24467. [PMID: 21912697 PMCID: PMC3166321 DOI: 10.1371/journal.pone.0024467] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 08/10/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Selective degeneration of medium spiny neurons and preservation of medium sized aspiny interneurons in striatum has been implicated in excitotoxicity and pathophysiology of Huntington's disease (HD). However, the molecular mechanism for the selective sparing of medium sized aspiny neurons and vulnerability of projection neurons is still elusive. The pathological characteristic of HD is an extensive reduction of the striatal mass, affecting caudate putamen. Somatostatin (SST) positive neurons are selectively spared in HD and Quinolinic acid/N-methyl-D-aspartic acid induced excitotoxicity, mimic the model of HD. SST plays neuroprotective role in excitotoxicity and the biological effects of SST are mediated by five somatostatin receptor subtypes (SSTR1-5). METHODS AND FINDINGS To delineate subtype selective biological responses we have here investigated changes in SSTR1 and 5 double knockout mice brain and compared with HD transgenic mouse model (R6/2). Our study revealed significant loss of dopamine and cAMP regulated phosphoprotein of 32 kDa (DARPP-32) and comparable changes in SST, N-methyl-D-aspartic acid receptors subtypes, calbindin and brain nitric oxide synthase expression as well as in key signaling proteins including calpain, phospho-extracellular-signal-regulated kinases1/2, synapsin-IIa, protein kinase C-α and calcineurin in SSTR1/5(-/-) and R6/2 mice. Conversely, the expression of somatostatin receptor subtypes, enkephalin and phosphatidylinositol 3-kinases were strain specific. SSTR1/5 appears to be important in regulating NMDARs, DARPP-32 and signaling molecules in similar fashion as seen in HD transgenic mice. CONCLUSIONS This is the first comprehensive description of disease related changes upon ablation of G- protein coupled receptor gene. Our results indicate that SST and SSTRs might play an important role in regulation of neurodegeneration and targeting this pathway can provide a novel insight in understanding the pathophysiology of Huntington's disease.
Collapse
Affiliation(s)
- Padmesh S. Rajput
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Geetanjali Kharmate
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Norman
- Department of Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shi-He Liu
- Department of Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bhagavatula R. Sastry
- Neuroscience Research Laboratory, Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Charles F. Brunicardi
- Department of Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ujendra Kumar
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
14
|
Gagnon J, Mayne J, Chen A, Raymond A, Woulfe J, Mbikay M, Chrétien M. PCSK2-null mice exhibit delayed intestinal motility, reduced refeeding response and altered plasma levels of several regulatory peptides. Life Sci 2011; 88:212-7. [DOI: 10.1016/j.lfs.2010.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 10/10/2010] [Accepted: 11/08/2010] [Indexed: 11/24/2022]
|
15
|
NOS1AP regulates dendrite patterning of hippocampal neurons through a carboxypeptidase E-mediated pathway. J Neurosci 2009; 29:8248-58. [PMID: 19553464 DOI: 10.1523/jneurosci.5287-08.2009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
During neuronal development, neurons form elaborate dendritic arbors that receive signals from axons. Additional studies are needed to elucidate the factors regulating the establishment of dendritic patterns. Our work explored possible roles played by nitric oxide synthase 1 adaptor protein (NOS1AP; also known as C-terminal PDZ ligand of neuronal nitric oxide synthase or CAPON) in dendritic patterning of cultured hippocampal neurons. Here we report that the long isoform of NOS1AP (NOS1AP-L) plays a novel role in regulating dendrite outgrowth and branching. NOS1AP-L decreases dendrite number when overexpressed at any interval between day in vitro (DIV) 0 and DIV 12, and knockdown of NOS1AP-L results in increased dendrite number. In contrast, the short isoform of NOS1AP (NOS1AP-S) decreases dendrite number only when overexpressed during DIV 5-7. Using mutants of NOS1AP-L, we show that neither the PDZ-binding domain nor the PTB domain is necessary for the effects of NOS1AP-L. We have functionally narrowed the region of NOS1AP-L that mediates this effect to the middle amino acids 181-307, a region that is not present in NOS1AP-S. Furthermore, we performed a yeast two-hybrid screen and identified carboxypeptidase E (CPE) as a binding partner for the middle region of NOS1AP-L. Biochemical and cellular studies reveal that CPE mediates the effects of NOS1AP on dendrite morphology. Together, our results suggest that NOS1AP-L plays an important role in the initiation, outgrowth, and maintenance of dendrites during development.
Collapse
|
16
|
Hakim AW, Dong XD, Svensson P, Kumar U, Cairns BE. TNFalpha mechanically sensitizes masseter muscle afferent fibers of male rats. J Neurophysiol 2009; 102:1551-9. [PMID: 19553487 DOI: 10.1152/jn.00326.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Behavioral evidence in rats indicates that injection of tumor necrosis factor alpha (TNFalpha) into skeletal muscle results in a prolonged mechanical sensitization without gross inflammation. To investigate whether a peripheral mechanism could underlie this effect, in the present study, TNFalpha (1 or 0.1 microg) was injected into the rat masseter muscle to assess its effect on the excitability and mechanical threshold (MT) of muscle nociceptors as well as on inflammation. Expression of TNFR1 (P55 receptors) and TNFR2 (P75 receptors) by the masseter muscle and trigeminal ganglion neurons that innervate that muscle was determined by Western blot and immunohistochemistry, respectively. The Evans blue dye technique was used at the end of the TNFalpha experiments to assess for plasma protein extravasation. In subsequent experiments to confirm the involvement of receptor activation in TNFalpha-induced effects, P55 or P75 receptor antibody was co-injected with TNFalpha. Intramuscular injection of 1 microg TNFalpha did not excite nociceptors but did significantly decrease MT compared with vehicle control. There was no evidence of gross inflammation 3 h after injection of TNFalpha. Co-injection of TNFalpha with P55 or P75 receptor antibodies attenuated TNFalpha-induced mechanical sensitization. P55 and P75 receptors were expressed by 29 and 62% of masseter nociceptors, respectively. These findings indicate that TNFalpha induces mechanical sensitization of masseter nociceptors that is mediated through activation of peripheral P55 and P75 receptors. These results support the hypothesis that a peripheral receptor mechanism could contribute to TNFalpha-induced noninflammatory mechanical sensitization of skeletal muscle previously reported in behaving rats.
Collapse
Affiliation(s)
- Akhlaq W Hakim
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, British Columbia, V6T 1Z3 Canada
| | | | | | | | | |
Collapse
|
17
|
Zhan S, Zhao H, J White A, Minami M, Pignataro G, Yang T, Zhu X, Lan J, Xiong Z, Steiner DF, Simon RP, Zhou A. Defective neuropeptide processing and ischemic brain injury: a study on proprotein convertase 2 and its substrate neuropeptide in ischemic brains. J Cereb Blood Flow Metab 2009; 29:698-706. [PMID: 19142196 PMCID: PMC3878611 DOI: 10.1038/jcbfm.2008.161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Using a focal cerebral ischemia model in rats, brain ischemia-induced changes in expression levels of mRNA and protein, and activities of proprotein convertase 2 (PC2) in the cortex were examined. In situ hybridization analyses revealed a transient upregulation of the mRNA level for PC2 at an early reperfusion hour, at which the level of PC2 protein was also high as determined by immunocytochemistry and western blotting. When enzymatic activities of PC2 were analyzed using a synthetic substrate, a significant decrease was observed at early reperfusion hours at which levels of PC2 protein were still high. Also decreased at these reperfusion hours were tissue levels of dynorphin-A(1-8) (DYN-A(1-8)), a PC2 substrate, as determined by radioimmunoassay. Further examination of PC2 protein biosynthesis by metabolic labeling in cultured neuronal cells showed that in ischemic cells, the proteolytic processing of PC2 was greatly attenuated. Finally, in mice, an intracerebroventricular administration of synthetic DYN-A(1-8) significantly reduced the extent of ischemic brain injury. In mice those lack an active PC2, exacerbated brain injury was observed after an otherwise non-lethal focal ischemia. We conclude that brain ischemia attenuates PC2 and PC2-mediated neuropeptide processing. This attenuation may play a role in the pathology of ischemic brain injury.
Collapse
Affiliation(s)
- Shuqin Zhan
- Robert S. Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon 97232, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Bakondi G, Pór Á, Kovács I, Szűcs G, Rusznák Z. Hyperpolarization-activated, cyclic nucleotide-gated, cation non-selective channel subunit expression pattern of guinea-pig spiral ganglion cells. Neuroscience 2009; 158:1469-77. [DOI: 10.1016/j.neuroscience.2008.10.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 10/29/2008] [Accepted: 10/30/2008] [Indexed: 11/26/2022]
|
19
|
Oiso S, Takeda Y, Futagawa T, Miura T, Kuchiiwa S, Nishida K, Ikeda R, Kariyazono H, Watanabe K, Yamada K. Contactin-associated protein (Caspr) 2 interacts with carboxypeptidase E in the CNS. J Neurochem 2009; 109:158-67. [PMID: 19166515 DOI: 10.1111/j.1471-4159.2009.05928.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To identify proteins interacting with the intracellular domain of the neural cell adhesion molecule contactin-associated protein 2 (Caspr2), yeast two-hybrid screening was performed. We identified carboxypeptidase E (CPE) as a Caspr2-interacting candidate protein. Glutathione S-transferase pull-down and immunoprecipitation analyses indicated that Caspr2 was associated with CPE in vitro and in vivo. Both Caspr2 and CPE were expressed predominantly in the CNS. Immunohistochemical analyses revealed that both Caspr2- and CPE-like immunoreactivities were found to co-localize in the apical dendrites and cell bodies of rat cortical neurons. In subcellular localization analysis, Caspr2- and CPE-like immunoreactivities were co-migrated in the fractions of Golgi/ER. Additionally, in COS-7 cells co-transfected with CPE and Caspr2 cDNAs, Caspr2- and CPE-immunoreactivities were co-localized in both Golgi and membrane, whereas it was only observed in Golgi of either COS-7 cell transfected with CPE or Caspr2 cDNA alone. It is known that the membrane-bound form of CPE functions as a sorting receptor of prohormones in the trans-Golgi network. Taken together, our data suggest that CPE may be a key molecule to regulate Caspr2 trafficking to the cell membrane.
Collapse
Affiliation(s)
- Shigeru Oiso
- Department of Clinical Pharmacy and Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|