Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Animals (Basel) ; 14(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38998005

ABSTRACT

Egg production traits are crucial in the poultry industry, including age at first egg (AFE), egg number (EN) at different stages, and laying rate (LR). Ducks exhibit higher egg production capacity than other poultry species, but the genetic mechanisms are still poorly understood. In this study, we collected egg-laying data of 618 Peking ducks from 22 to 66 weeks of age and genotyped them by whole-genome resequencing. Genetic parameters were calculated based on SNPs, and a genome-wide association study (GWAS) was performed for these traits. The SNP-based heritability of egg production traits ranged from 0.09 to 0.54. The GWAS identified nine significant SNP loci associated with AFE and egg number from 22 to 66 weeks. These loci showed that the corresponding alleles were positively correlated with a decrease in the traits. Moreover, three potential candidate genes (ENSAPLG00020011445, ENSAPLG00020012564, TMEM260) were identified. Functional enrichment analyses suggest that specific immune responses may have a critical impact on egg production capacity by influencing ovarian function and oocyte maturation processes. In conclusion, this study deepens the understanding of egg-laying genetics in Peking duck and provides a sound theoretical basis for future genetic improvement and genomic selection strategies in poultry.

2.
Mol Psychiatry ; 26(9): 4605-4615, 2021 09.
Article in English | MEDLINE | ID: mdl-32504049

ABSTRACT

Evidence from epidemiological and laboratory studies, as well as randomized placebo-controlled trials, suggests supplementation with n-3 polyunsaturated fatty acids (PUFAs) may be efficacious for treatment of major depressive disorder (MDD). The mechanisms underlying n-3 PUFAs potential therapeutic properties remain unknown. There are suggestions in the literature that glial hypofunction is associated with depressive symptoms and that antidepressants may normalize glial function. In this study, induced pluripotent stem cells (iPSC)-derived neuronal stem cell lines were generated from individuals with MDD. Astrocytes differentiated from patient-derived neuronal stem cells (iNSCs) were verified by GFAP. Cells were treated with eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) or stearic acid (SA). During astrocyte differentiation, we found that n-3 PUFAs increased GFAP expression and GFAP positive cell formation. BDNF and GDNF production were increased in the astrocytes derived from patients subsequent to n-3 PUFA treatment. Stearic Acid (SA) treatment did not have this effect. CREB activity (phosphorylated CREB) was also increased by DHA and EPA but not by SA. Furthermore, when these astrocytes were treated with n-3 PUFAs, the cAMP antagonist, RP-cAMPs did not block n-3 PUFA CREB activation. However, the CREB specific inhibitor (666-15) diminished BDNF and GDNF production induced by n-3 PUFA, suggesting CREB dependence. Together, these results suggested that n-3 PUFAs facilitate astrocyte differentiation, and may mimic effects of some antidepressants by increasing production of neurotrophic factors. The CREB-dependence and cAMP independence of this process suggests a manner in which n-3 PUFA could augment antidepressant effects. These data also suggest a role for astrocytes in both MDD and antidepressant action.


Subject(s)
Depressive Disorder, Major , Fatty Acids, Omega-3 , Neural Stem Cells , Astrocytes , Docosahexaenoic Acids/pharmacology , Fatty Acids, Omega-3/pharmacology , Humans , Nerve Growth Factors , Neurogenesis
3.
Am J Clin Pathol ; 152(5): 647-655, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31305883

ABSTRACT

OBJECTIVES: To investigate the diagnostic potential of AEG-1 and GPC-3 in hepatocellular carcinoma (HCC). METHODS: AEG-1 and GPC-3 immunohistochemistry were performed on HCC, adjacent nontumor tissue (ANT), and dysplastic nodules (DN). RESULTS: H score of AEG-1 or GPC-3 in HCC was significantly higher than in ANT or DN. In HCC, 92% and 54% showed AEG-1 and GPC-3 positivity, respectively. In ANT, 16.2% were AEG-1 and 7.6% GPC-3 positive. AEG-1 staining was mostly diffuse, whereas GPC-3 frequently showed focal staining. AEG-1 alone showed high sensitivity but low specificity and accuracy. GPC-3, on the other hand, showed high specificity but low sensitivity and accuracy. Combination of both stains boosted the sensitivity, specificity, and accuracy to 94.6%, 89.5%, and 90.5%, respectively, when only diffuse staining was considered as positive. CONCLUSIONS: AEG-1 or GPC-3 alone seemed not an ideal marker for HCC. The combination of AEG-1 and GPC-3 might improve early diagnosis of HCC.


Subject(s)
Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/diagnosis , Glypicans/analysis , Liver Neoplasms/chemistry , Liver Neoplasms/diagnosis , Membrane Proteins/analysis , RNA-Binding Proteins/analysis , Aged , Biomarkers, Tumor/analysis , Female , Gene Expression , Glypicans/genetics , Humans , Immunohistochemistry , Male , Membrane Proteins/genetics , Middle Aged , RNA-Binding Proteins/genetics , Sensitivity and Specificity
4.
Radiat Res ; 191(1): 107-121, 2019 01.
Article in English | MEDLINE | ID: mdl-30430918

ABSTRACT

The development of effective biomarkers for detecting the magnitude of radiation exposure and resiliency of host response is crucial to identifying appropriate treatment strategies after radiation exposure. We hypothesized that the gastrointestinal resident bacteria would demonstrate predictable, dose-dependent changes after radiation exposure across two large animal models of acute radiation syndrome. Here, Göttingen minipigs (GMP) (n = 50) and rhesus macaques (n = 48) were exposed to five dose levels (resulting in mortality rates of 33-100% and 25-68.7%, respectively). Fecal samples taken prior to and after irradiation (day 0 for GMP; day 0, 3 and 14 for macaques) were used for 16S rRNA gene sequence amplicon high-throughput sequencing. Baseline gut microbiota profiles were dissimilar between GMP and macaques, however, radiation appeared to have similar effect at the phylum level, resulting in Bacteroidetes decrease and Firmicutes increase in both models. The abundance of the main Bacteroidetes genus ( Bacteroides for GMP, Prevotella for macaques) was profoundly decreased by irradiation. Intracellular symbionts [Elusimicrobia in GMP, Treponema (Spirochaetes) in macaques] consistently increased after irradiation, suggesting their use as potential biomarkers of intestinal injury, and potential negative effect on health. Prevotella, Lactobacillus, Clostridium XIVa, Oscillibacter and Elusimicrobium/ Treponema abundances were found to be very significantly correlated with radiation intensity. Furthermore, Prevotella, Enterorhabdus and Ruminococcus and Enterorhabdus maintenance was strongly associated with survival in GMP, while Prevotella, Oscillibacter and Treponema were strongly associated with survival and Streptococcus with death in macaques. Overall, we found that a wide range of gut bacterial genera known to be abundant in the human gut microbiota are excellent biomarkers of radiation intensity and resilience in animal models, and that detrimental effects can be monitored, and potentially prevented, by targeting selected genera.


Subject(s)
Acute Radiation Syndrome/mortality , Gastrointestinal Microbiome , Models, Animal , Radiation Dosage , Acute Radiation Syndrome/etiology , Animals , Biomarkers/metabolism , High-Throughput Nucleotide Sequencing , Humans , Macaca mulatta , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Swine , Swine, Miniature
5.
Radiat Environ Biophys ; 57(4): 419-426, 2018 11.
Article in English | MEDLINE | ID: mdl-30343431

ABSTRACT

In rodent studies, the gut microbiota has been implicated in facilitating both radioresistance, by protecting the epithelium from apoptotic responses and radiosensitivity, inducing endothelial apoptotic responses. Despite the observation that large animal models, such as the Chinese Rhesus macaque and the Gottingen Minipig, demonstrate similarity to human physiologic responses to radiation, little is known about radiation-induced changes of the gut microbiome in these models. To compare the two models, we used bioequivalent radiation doses which resulted in an LD50 for Gottingen Minipigs and Chinese Rhesus macaques, 1.9 Gy and 6.8 Gy, respectively. Fecal samples taken prior and 3 days post-radiation were used for 16S rRNA gene sequence amplicon high throughput sequencing (Illumina MiSeq). Baseline gut microbiota profiles were dissimilar between minipigs and rhesus macaques. Irradiation profoundly impacted gut microbiota profiles in both animals. Significant increases of intracellular symbionts were common to both models and to reported changes in rodents suggesting universality of these findings post-radiation. Remarkably, opposite dynamics were observed for the main phyla, with increase of Firmicutes and decrease of Bacteroidetes and Proteobacteria in minipigs but with enrichment of Bacteroidetes in rhesus macaques. Minipig changes in magnitude and in variety of species affected were more extensive than those observed in rhesus macaques. This pilot study provides an important first step in comparing the radiosensitive pig model to the comparatively more radioresistant macaque model, for the identification of microbial elements which may influence radiosensitivity.


Subject(s)
Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/microbiology , Gastrointestinal Microbiome/radiation effects , Radiation Exposure/adverse effects , Animals , Disease Models, Animal , Kaplan-Meier Estimate , Macaca mulatta , Swine , Swine, Miniature , Therapeutic Equivalency
7.
Photomed Laser Surg ; 34(11): 556-563, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27244220

ABSTRACT

OBJECTIVE: To address whether a single treatment of one of three visible light wavelengths, 635, 532, and 405 nm (constant wave, energy density 2.9 J/m2), could affect the hallmarks of established renal fibrosis and whether these wavelengths could facilitate mesenchymal stem cell (MSC) beneficence. BACKGROUND DATA: Chronic kidney disease is a global health problem with only 20% receiving care worldwide. Kidneys with compromised function have ongoing inflammation, including increased oxidative stress and apoptosis, peritubular capillary loss, tubular atrophy, and tubulointerstitial fibrosis. Promising studies have highlighted the significant potential of MSC-based strategies to mitigate fibrosis; however, reversal of established fibrosis has been problematic, suggesting that methods to potentiate MSC effects require further development. Laser treatments at visible wavelengths have been reported to enhance mitochondrial potential and available cellular ATP, facilitate proliferation, and inhibit apoptosis. We hypothesized that laser-delivered energy might provide wavelength-specific effects in the fibrotic kidney and enhance MSC responses. MATERIALS AND METHODS: Renal fibrosis, established in C57BL6 mice following 21 days of unilateral ureter obstruction (UUO), was treated with one of three wavelengths alone or with autologous MSC. Mitochondrial activity, cell proliferation, apoptosis, and cytokines were measured 24 h later. RESULTS: Wavelengths 405, 532, and 635 nm all significantly synergized with MSC to enhance mitochondrial activity and reduce apoptosis. Proliferative activity was observed in the renal cortices following combined treatment with the 532 nm laser and MSC; endothelial proliferation increased in response to the 635 nm laser alone and to the combined effects of MSC and the 405 nm wavelength. Reductions of transforming growth factor-ß were observed with 532 nm alone and when combined with MSC. CONCLUSIONS: Specific wavelengths of laser energy appear to induce different responses in renal fibrotic tissue. These findings support further study in the development of a customized laser therapy program of combined wavelengths to optimize MSC effects in the treatment of renal fibrosis.


Subject(s)
Fibrosis/radiotherapy , Kidney Diseases/radiotherapy , Low-Level Light Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Regeneration/radiation effects , Animals , Apoptosis/radiation effects , Biopsy, Needle , Disease Models, Animal , Fibrosis/pathology , Fibrosis/surgery , Fluorescent Antibody Technique , Immunohistochemistry , Kidney Diseases/pathology , Kidney Diseases/surgery , Lasers , Male , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Inbred C57BL , Pilot Projects , Random Allocation , Reference Values , Regeneration/physiology , Transplantation, Autologous
8.
BMC Cancer ; 15: 915, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26582057

ABSTRACT

BACKGROUND: There is limited information about the clinical and biological significance of prostate specific G protein coupled receptor (PSGR) in prostate cancer (PCa) initiation and progression. Here, we evaluated the expression of PSGR protein, studied its diagnostic and prognostic value in PCa, and also explored its role in cancer cell growth and invasion. METHODS: The expression of PSGR in paired adjacent normal prostate, high grade prostatic intraepithelial neoplasia (PIN), and PCa were determined by immunohistochemistry on tissue microarrays constructed from 150 radical prostatectomy specimens. The effects of PSGR on PCa cell growth and invasion were investigated using human PCa cell lines. RESULTS: Membranous and cytoplasmic PSGR staining was observed at luminal epithelial cells of prostate. PSGR protein expression was significantly higher in PIN compared to normal prostate. Interestingly, the expression of PSGR decreased as PIN progressed to PCa. Low PSGR expression in PCa was associated with high Gleason score, and poor overall survival. Activated PSGR increased cancer cell invasive ability, but retarded cell growth. PSGR did not affect mTOR activity, but suppressed P70 S6 kinase activity. CONCLUSIONS: PSGR may participate in PCa progression through affecting cell proliferation and invasion. High expression of PSGR in PIN may implicate its role in early neoplastic transformation of PCa. Low expression of PSGR in PCa may serve as a potential indicator for poor prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Adult , Aged , Cell Membrane/metabolism , Cell Proliferation/physiology , Cytoplasm/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Prognosis , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/diagnosis , Survival Analysis , Tissue Array Analysis , Tumor Cells, Cultured
9.
Br J Cancer ; 113(5): 738-46, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26263481

ABSTRACT

BACKGROUND: Currently, there is a lack of ideal biomarkers for predicting nodal status in preoperative stage of oesophageal adenocarcinoma (EAC) to aid optimising therapeutic options. We studied the potential of applying subtype macrophages to predict lymph node metastasis and prognosis in EAC. MATERIAL AND METHODS: Fifty-three EAC resection specimens were immunostained with CD68, CD40 (M1), and CD163 (M2). Lymphatic vessel density (LVD) was estimated with the staining of D2-40. Subsequently, we tested if M2d macrophage could promote EAC cell migration and invasion. RESULTS: In EAC without neoadjuvant treatment, an increase in M2-like macrophage was associated with poor patient survival, independent of the locations of macrophages in tumour. The M2/M1 ratio that represented the balance between M2- and M1-like macrophages was significantly higher in nodal-positive EACs than that in nodal-negative EACs, and inversely correlated with patient overall survival. The M2/M1 ratio was not related to LVD. EAC cell polarised THP1 cell into M2d-like macrophage, which promoted EAC cell migration and invasion. Neoadjuvant therapy appeared to diminish the correlation between the M2/M1 ratio and survival. CONCLUSIONS: The ratio of M2/M1 macrophage may serve as a sensitive marker to predict lymph node metastasis and poor prognosis in EAC without neoadjuvant therapy. M2d macrophage may have important roles in EAC metastasis.


Subject(s)
Adenocarcinoma/secondary , Esophageal Neoplasms/pathology , Macrophages/immunology , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adult , Aged , Cell Line, Tumor , Cell Movement , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Female , Humans , Kaplan-Meier Estimate , Lymph Nodes/pathology , Lymphatic Metastasis , Lymphatic Vessels/pathology , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Invasiveness , Young Adult
10.
Radiat Res ; 184(1): 46-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26121229

ABSTRACT

In a mass casualty radiation event situation, individualized therapy may overwhelm available resources and feasibility issues suggest a need for the development of population-based strategies. To investigate the efficacy of a population-based strategy, Chinese macaques (n = 46) underwent total-body irradiation and received preemptive antibiotics, IV hydration on predetermined postirradiation days and were then compared to macaques (n = 48) that received subject-based care in which blood transfusions, IV hydration, nutritional supplementation and antibiotic supportive measures were provided. Estimated radiation doses for LD30/60, LD50/60 and LD70/60 of animals with subject-based care: 6.83 Gy (6.21, 7.59), 7.44 Gy (6.99, 7.88) and 8.05 Gy (7.46, 8.64), respectively, and for population-based care: 5.61 Gy (5.28, 6.17), 6.62 Gy (6.13, 7.18) and 7.63 Gy (7.21, 8.20), respectively. Analysis of four time periods, 0-9, 10-15, 16-25 and 26-60 days postirradiation, identified significant mortality differences during the period of 10-15 days. A subset analysis of higher radiation doses (6.75-7.20 Gy, n = 32) indicated hydration, nutrition and septic status were not significantly different between treatments. Whole blood transfusion treatment, administered only in subject-supportive care, was associated with significantly higher platelet and absolute neutrophil counts. Median platelet counts greater than 5,670 cells/µl and absolute neutrophil counts greater than 26 cells/µl during this period correlated with survival. We observed that the population-based treatment increased the LD50/60 compared to nontreatment (6.62 Gy vs. 4.92 Gy) and may be further optimized during days 10-15, where strategic blood transfusions or other strategies to achieve increases in neutrophil and platelet counts may further increase survival rates in subjects exposed to high doses of radiation.


Subject(s)
Radiation Injuries, Experimental/therapy , Animals , Anti-Bacterial Agents/therapeutic use , Blood Transfusion , Macaca mulatta , Male , Neutropenia/therapy , Nutritional Support , Thrombocytopenia/therapy , Whole-Body Irradiation
11.
Inflamm Bowel Dis ; 20(4): 703-11, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24518605

ABSTRACT

BACKGROUND: Metastasis-associated in colon cancer-1 (MACC1), a newly identified regulator of HGF-MET signaling, may participate into the key steps of sporadic colorectal adenocarcinoma development. Given there are many pathogenetic distinctions between colitis-associated colorectal cancer (CAC) and sporadic colorectal adenocarcinomas, the potential roles of MACC1 in CAC carcinogenesis remain unknown. For the first time, we evaluated the expressions of MACC1 and MET in IBD-associated colitis, dysplasia, and adenocarcinoma. METHODS: Expression was investigated by immunohistochemistry in tissue microarrays consisting of 13 normal colon, 11 active colitis, 9 dysplasia, 51 conventional CAC, 5 mucinous adenocarcinoma, and 1 signet ring cell adenocarcinoma specimens. The expression level of MACC1 or MET was evaluated with H-score system. RESULTS: MACC1 expression was significantly higher in IBD-associated dysplasia than that in corresponding inflammatory or normal colonic tissue, and its level was further elevated from dysplasia to conventional CAC. Higher MACC1 expression was seen in a patient with CAC who had multifocal dysplasia or synchronous carcinoma. MACC1 overexpression (H-score >100) was seen in 67% of conventional CAC but in 0% of dysplasia and 0% of inflammation or normal colon. There was no difference of MACC1 expression found among well, moderately and poorly differentiated CAC. MET expressions in inflammation, dysplasia, and conventional CAC were statistically similar. No parallel expression of MACC1 and MET was detected in this study. MACC1 and MET expression was not increased in mucinous or signet ring cell carcinoma, 2 distinct variants of CAC. CONCLUSIONS: Stepwise increase of MACC1 expression from IBD-associated colitis to dysplasia to adenocarcinoma suggests that MACC1 is strongly associated with conventional CAC tumorigenesis in a manner independent of MET. MACC1 may serve as a potential marker for early diagnosis of conventional CAC.


Subject(s)
Adenocarcinoma/chemistry , Colitis, Ulcerative/metabolism , Colorectal Neoplasms/chemistry , Crohn Disease/metabolism , Proto-Oncogene Proteins c-met/analysis , Transcription Factors/analysis , Adenocarcinoma/pathology , Adenocarcinoma, Mucinous/chemistry , Adult , Aged , Carcinoma, Signet Ring Cell/chemistry , Colitis, Ulcerative/pathology , Colon/chemistry , Colorectal Neoplasms/pathology , Crohn Disease/pathology , Female , Humans , Male , Middle Aged , Neoplasm Grading , Proto-Oncogene Proteins c-met/metabolism , Trans-Activators , Transcription Factors/metabolism , Young Adult
12.
Am J Clin Pathol ; 140(5): 701-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24124150

ABSTRACT

OBJECTIVES: To investigate metastasis associated in colon cancer 1 (MACC1) and MET expression in colorectal adenoma, Tis, early-stage invasive (T1 and T2), and advanced adenocarcinoma with liver metastasis using immunohistochemistry. METHODS: Ninety-three paraffin-embedded colorectal tumor specimens were immunohistochemically analyzed for MACC1 and MET protein expression. RESULTS: MACC1 expression was upregulated in the transition from adenoma to Tis; its expression was further elevated during tumor progression from Tis to early invasive carcinoma. MET expression was constant from adenoma to Tis and to T1 but significantly increased as tumor progression to T2. Both MACC1 and MET expression were enhanced in advanced carcinoma with liver metastasis. CONCLUSIONS: Stepwise elevation of MACC1 expression in key points of colorectal cancer development suggests that MACC1 may contribute to cancer initiation and early invasive growth. High expression of both MACC1 and MET may relate to distant metastasis.


Subject(s)
Adenocarcinoma/secondary , Adenoma/pathology , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Transcription Factors/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Disease Progression , Early Diagnosis , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Trans-Activators , Transcription Factors/metabolism , Up-Regulation
13.
Mol Endocrinol ; 27(1): 38-49, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23204329

ABSTRACT

Estrogen signaling, through estrogen receptor (ER)α, has been shown to cause hypertrophy in the prostate. Our recent report has shown that epithelial ERα knockout (KO) will not affect the normal prostate development or homeostasis. However, it remains unclear whether ERα in different types of stromal cells has distinct roles in prostate development. This study proposed to elucidate how KO of ERα in the stromal smooth muscle or fibroblast cells may interrupt cross talk between prostate stromal and epithelial cells. Smooth muscle ERαKO (smERαKO) mice showed decreased glandular infolding with the proximal area exhibiting a significant decrease. Fibroblast ERαKO mouse prostates did not exhibit this phenotype but showed a decrease in the number of ductal tips. Additionally, the amount of collagen observed in the basement membrane was reduced in smERαKO prostates. Interestingly, these phenotypes were found to be mutually exclusive among smERαKO or fibroblast ERαKO mice. Compound KO of ERα in both fibroblast and smooth muscle showed combined phenotypes from each of the single KO. Further mechanistic studies showed that IGF-I and epidermal growth factor were down-regulated in prostate smooth muscle PS-1 cells lacking ERα. Together, our results indicate the distinct functions of fibroblast vs. smERα in prostate development.


Subject(s)
Estrogen Receptor alpha/metabolism , Fibroblasts/metabolism , Myocytes, Smooth Muscle/metabolism , Prostate/growth & development , Animals , Basement Membrane/metabolism , Cell Line , Collagen/metabolism , Epidermal Growth Factor/physiology , Estradiol/physiology , Estrogen Receptor alpha/genetics , Female , Gene Knockout Techniques , Homeostasis , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis , Prostate/cytology , Prostate/metabolism , Somatomedins/physiology
14.
Handb Clin Neurol ; 106: 9-35, 2012.
Article in English | MEDLINE | ID: mdl-22608613

ABSTRACT

This volume describes a series of psychiatric and neuropsychiatric disorders, connects some aspects of somatic and psychiatric medicine, and describes various current and emerging therapies. The purpose of this chapter is to set the stage for the volume by developing the theoretical basis of synaptic transmission and introducing the various neurotransmitters and their receptors involved in the process. The intent is to provide not only a historical context through which to understand neurotransmitters, but a current contextual basis for understanding neuronal signal transduction and applying this knowledge to facilitate treatment of maladies of the brain and mind.


Subject(s)
Receptors, Cell Surface/physiology , Synaptic Transmission/physiology , Animals , Cell Biology , Humans , Ligands , Models, Biological , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Cell Surface/drug effects , Synaptic Transmission/drug effects
15.
PLoS One ; 7(12): e52838, 2012.
Article in English | MEDLINE | ID: mdl-23285198

ABSTRACT

Although evidence has shown the regulating effect of n-3 poly-unsaturated fatty acid (n-3 PUFA) on cell signaling transduction, it remains unknown whether n-3 PUFA treatment modulates estrogen signaling. The current study showed that docosahexaenoic acid (DHA, C22:6), eicosapentaenoic acid (EPA, C20:5) shifted the pro-survival and proliferative effect of estrogen to a pro-apoptotic effect in human breast cancer (BCa) MCF-7 and T47D cells. 17 ß-estradiol (E2) enhanced the inhibitory effect of n-3 PUFAs on BCa cell growth. The IC50 of DHA or EPA in MCF-7 cells decreased when combined with E2 (10 nM) treatment (from 173 µM for DHA only to 113 µM for DHA+E2, and from 187 µm for EPA only to 130 µm for EPA+E2). E2 also augmented apoptosis in n-3 PUFA-treated BCa cells. In contrast, in cells treated with stearic acid (SA, C18:0) as well as cells not treated with fatty acid, E2 promoted breast cancer cell growth. Classical (nuclear) estrogen receptors may not be involved in the pro-apoptotic effects of E2 on the n-3 PUFA-treated BCa cells because ERα agonist failed to elicit, and ERα knockdown failed to block E2 pro-apoptotic effects. Subsequent studies reveal that G protein coupled estrogen receptor 1 (GPER1) may mediate the pro-apoptotic effect of estrogen. N-3 PUFA treatment initiated the pro-apoptotic signaling of estrogen by increasing GPER1-cAMP-PKA signaling response, and blunting EGFR, Erk 1/2, and AKT activity. These findings may not only provide the evidence to link n-3 PUFAs biologic effects and the pro-apoptotic signaling of estrogen in breast cancer cells, but also shed new insight into the potential application of n-3 PUFAs in BCa treatment.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Fatty Acids, Omega-3/metabolism , Signal Transduction , Apoptosis/drug effects , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , ErbB Receptors/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Fatty Acids, Omega-3/pharmacology , Female , Humans , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
16.
Neurosignals ; 17(1): 100-8, 2009.
Article in English | MEDLINE | ID: mdl-19212143

ABSTRACT

A large percentage of current drugs target G-protein-coupled receptors, which couple to well-known signaling pathways involving cAMP or calcium. G-proteins themselves may subserve a second messenger function. Here, we review the role of tubulin and microtubules in directly mediating effects of heterotrimeric G-proteins on neuronal outgrowth, shape and differentiation. G-protein-tubulin interactions appear to be regulated by neurotransmitter activity, and, in turn, regulate the location of Galpha in membrane microdomains (such as lipid rafts) or the cytosol. Tubulin binds with nanomolar affinity to Gsalpha, Gialpha1 and Gqalpha (but not other Galpha subunits) as well as Gbeta(1)gamma(2) subunits. Galpha subunits destabilize microtubules by stimulating tubulin's GTPase, while Gbetagamma subunits promote microtubule stability. The same region on Gsalpha that binds adenylyl cyclase and Gbetagamma also interacts with tubulin, suggesting that cytoskeletal proteins are novel Galpha effectors. Additionally, intracellular Gialpha-GDP, in concert with other GTPase proteins and Gbetagamma, regulates the position of the mitotic spindle in mitosis. Thus, G-protein activation modulates cell growth and differentiation by directly altering microtubule stability. Further studies are needed to fully establish a structural mechanism of this interaction and its role in synaptic plasticity.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Microtubules/metabolism , Tubulin/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Membrane/metabolism , Cytosol/metabolism , GTP Phosphohydrolases/metabolism , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Models, Molecular , Neurogenesis , Neurons/physiology , Neurons/ultrastructure , Neurotransmitter Agents/metabolism , Protein Stability , Protein Structure, Quaternary , Spindle Apparatus/physiology
17.
J Biol Chem ; 284(16): 10462-72, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19237344

ABSTRACT

It is now evident that Galpha(s) traffics into cytosol following G protein-coupled receptor activation, and alpha subunits of some heterotrimeric G-proteins, including Galpha(s) bind to tubulin in vitro. Nevertheless, many features of G-protein-microtubule interaction and possible intracellular effects of G protein alpha subunits remain unclear. In this study, several biochemical approaches demonstrated that activated Galpha(s) directly bound to tubulin and cellular microtubules, and fluorescence microscopy showed that cholera toxin-activated Galpha(s) colocalized with microtubules. The activated, GTP-bound, Galpha(s) mimicked tubulin in serving as a GTPase activator for beta-tubulin. As a result, activated Galpha(s) made microtubules more dynamic, both in vitro and in cells, decreasing the pool of insoluble microtubules without changing total cellular tubulin content. The amount of acetylated tubulin (an indicator of microtubule stability) was reduced in the presence of Galpha(s) activated by mutation. Previous studies showed that cholera toxin and cAMP analogs may stimulate neurite outgrowth in PC12 cells. However, in this study, overexpression of a constitutively activated Galpha(s) or activation of Galpha(s) with cholera toxin in protein kinase A-deficient PC12 cells promoted neurite outgrowth in a cAMP-independent manner. Thus, it is suggested that activated Galpha(s) acts as an intracellular messenger to regulate directly microtubule dynamics and promote neurite outgrowth. These data serve to link G-protein signaling with modulation of the cytoskeleton and cell morphology.


Subject(s)
Cytosol/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Microtubules/metabolism , Neurites/metabolism , Protein Isoforms/metabolism , Animals , Cholera Toxin/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein alpha Subunits/genetics , Guanosine Triphosphate/metabolism , Neurites/ultrastructure , PC12 Cells , Protein Isoforms/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/physiology , Tubulin/metabolism
18.
Cell Signal ; 19(11): 2247-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17692506

ABSTRACT

Mastoparan, a wasp venom toxin, has various pharmacological activities, the mechanisms of which are still unknown. To clarify the action of mastoparan on G protein-coupled receptor-mediated signaling, we previously examined the effect of mastoparan on G(q)-mediated signaling and demonstrated that mastoparan binds to gangliosides causing a decrease in Galpha(q/11) content in lipid rafts, and resulting in the inhibition of G(q)-mediated phosphoinositide hydrolysis (Sugama et al., Mol. Pharmacol., 68, 1466, 2005). In the present study, we examined the effect of mastoparan on beta-adrenoceptor-G(s) signaling in 1321N1 human astrocytoma cells. Mastoparan inhibited isoproterenol-induced elevation of cyclic AMP in a concentration-dependent manner. Although mastoparan is known to be an activator of G(i), pertussis toxin only slightly attenuated mastoparan-induced inhibition of cyclic AMP elevation, suggesting that a major part of the inhibition of cyclic AMP elevation induced by mastoparan is not mediated by Galpha(i). By contrast, mastoparan-induced inhibition of cyclic AMP elevation was clearly attenuated by preincubation of the cells with ganglioside mixtures. Moreover, mastoparan changed the localization of Galpha(s) in lipid rafts without disrupting the structure of lipid rafts. Fluorescent staining analysis showed that mastoparan released GFP-Galpha(s) from plasma membranes into the cytosol. These results suggest that the mastoparan-induced suppression of cyclic AMP elevation is mainly caused by changing the localization of Galpha(s) in lipid rafts into a compartment in the cellular interior where it is not available to activate adenylyl cyclase.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Peptides/pharmacology , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Wasp Venoms/pharmacology , Cell Line, Tumor , Cyclic AMP/biosynthesis , Cytosol/drug effects , Cytosol/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits/metabolism , Green Fluorescent Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Isoproterenol/pharmacology , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/drug effects
19.
FASEB J ; 20(9): 1452-61, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816120

ABSTRACT

The microtubule-associated protein tau may be involved in cell morphogenesis and axonal maintenance. In addition to microtubules, tau has been shown to interact with actin in vitro. In the present study interaction of tau and actin was investigated in PC12 cells. No interaction between tau and actin was observed without NGF treatment. Under NGF stimulation, tau distributed at ends of cellular extensions, where it associated with actin in a microtubule-independent manner. F-actin disruption revealed that relocalization and assembly of F-actin at the ends of cellular extensions were necessary for NGF-induced tau reorganization and association with actin. A truncated tau-GFP (tau(1-186)-GFP, N-terminal of tau) did not associate with actin. However, tau23(174-352)-GFP (carboxyl-terminal of Tau23) did associate with actin and the requirement for NGF was lost. Nevertheless, NGF boosted tau23(174-352)-GFP interaction with actin and promoted colocalization at the ends of cellular extensions. This suggests that the C-terminal of tau is required for associating with actin and the tau N-terminal may play a regulatory role in this process. A possible role for tau-actin interaction in neurite outgrowth is postulated.


Subject(s)
Actins/metabolism , tau Proteins/metabolism , Actins/genetics , Animals , Cell Differentiation , Genes, Reporter , Green Fluorescent Proteins/metabolism , PC12 Cells , Pheochromocytoma , Rats , Recombinant Fusion Proteins/metabolism , Transfection , tau Proteins/genetics
20.
J Cell Sci ; 117(Pt 25): 6129-41, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15564376

ABSTRACT

Tau is a major microtubule-associated protein which induces bundling and stabilization of axonal microtubules (MTs). To investigate the interaction of tau with MTs in living cells, we expressed GFP-tau fusion protein in cultured Xenopus embryo neurons and performed time-lapse imaging of tau-labeled MTs. Tau uniformly labeled individual MTs regardless of their assembly/disassembly status and location along the axon. Photobleaching experiments indicated that interaction of tau with MTs is very dynamic, with a half-time of fluorescence recovery of the order of 3 seconds. Treatment of cells with taxol, a drug that suppresses MT dynamics, rapidly induced detachment of tau from MTs. Although binding of tau to straight MTs was uniform, there was a heightened concentration of tau at the sites of high MT curvature. Our results suggest that dynamic interaction of tau with MTs may modify local mechanical properties of individual MTs and play a crucial role in the remodeling of the MT cytoskeleton during neuronal plasticity.


Subject(s)
Microtubules/metabolism , tau Proteins/metabolism , Animals , Axons/metabolism , Cytoskeleton/metabolism , DNA/metabolism , Detergents/pharmacology , Embryo, Nonmammalian/metabolism , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Image Processing, Computer-Assisted , Light , Microscopy, Fluorescence , Microtubules/drug effects , Microtubules/ultrastructure , Neurons/metabolism , Paclitaxel/pharmacology , Protein Binding , Time Factors , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...