Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Arch Biochem Biophys ; 730: 109396, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36113626

ABSTRACT

Preeclampsia (PE), a systemic vascular disorder, is the leading cause of maternal and perinatal morbidity and mortality, and its pathogenesis has yet to be fully elucidated. Siglec6, a transmembrane protein, is highly expressed in human placental trophoblasts, and previous studies have shown that Siglec6 overexpression correlates with PE, but the role of Siglec6 during PE progression is unknown. Here, we demonstrated that the mRNA and protein expression levels of Siglec6 were upregulated in early-onset PE placentas compared with uncomplicated pregnancies, and Siglec6 was primarily located in syncytiotrophoblasts (STBs) and extravillous trophoblasts (EVTs). Moreover, our results showed that chemical reagent-induced HIF-1α accumulation promoted the mRNA and protein levels of Siglec6 in HTR8/SVneo and BeWo cells. Although Siglec6 overexpression did not affect HTR8/SVneo cell proliferation, migration, and invasion, the conditional medium derived from the Siglec6 overexpressed HTR8/SVneo cells (Siglec6-OE-CM) significantly impaired the proliferation, migration, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs). Subsequently, the transcriptome sequencing results revealed that Siglec6 overexpression led to the downregulation of Wnt6 in HTR8/SVneo cells, which was further confirmed by qPCR and ELISA. Recombinant human Wnt6 reversed Siglec6-OE-CM-mediated suppression of HUVEC functions by reactivating the Wnt/ß-catenin signaling pathway. Altogether, our study found that elevated trophoblastic Siglec6 contributed to the impairment of vascular endothelial cell functions by downregulating Wnt6/ß-catenin signaling.


Subject(s)
Antigens, Differentiation, Myelomonocytic , Lectins , Pre-Eclampsia , Trophoblasts , Female , Humans , Pregnancy , beta Catenin/metabolism , Cell Line , Cell Movement , Cell Proliferation , Human Umbilical Vein Endothelial Cells/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , RNA, Messenger/metabolism , Trophoblasts/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, CD , Lectins/genetics
2.
Clin Sci (Lond) ; 136(4): 257-272, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35103285

ABSTRACT

Maternal circulating levels of the adipokine chemerin are elevated in preeclampsia, but its origin and contribution to preeclampsia remain unknown. We therefore studied (1) placental chemerin expression and release in human pregnancy, and (2) the consequences of chemerin overexpression via lentivirus-mediated trophoblast-specific gene manipulation in both mice and immortalized human trophoblasts. Placental chemerin expression and release were increased in women with preeclampsia, and their circulating chemerin levels correlated positively with the soluble Fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) ratio, a well-known biomarker of preeclampsia severity. Placental trophoblast chemerin overexpression in mice induced a preeclampsia-like syndrome, involving hypertension, proteinuria, and endotheliosis, combined with diminished trophoblast invasion, a disorganized labyrinth layer, and up-regulation of sFlt-1 and the inflammation markers nuclear factor-κB (NFκB), tumor necrosis factor (TNF)-α, and interleukin (IL)-1ß. It also led to embryo resorption, while maternal serum chemerin levels correlated negatively with fetal weight in mice. Chemerin overexpression in human trophoblasts up-regulated sFlt-1, reduced vascular endothelial factor-A, and inhibited migration and invasion, as well as tube formation during co-culture with human umbilical vein endothelial cells (HUVECs). The chemokine-like receptor 1 (CMKLR1) antagonist α-NETA prevented the latter phenomenon, although it did not reverse the chemerin-induced down-regulation of the phosphoinositide 3-kinase/Akt pathway. In conclusion, up-regulation of placental chemerin synthesis disturbs normal placental development via its CMKLR1 receptor, thereby contributing to fetal growth restriction/resorption and the development of preeclampsia. Chemerin might be a novel biomarker of preeclampsia, and inhibition of the chemerin/CMKLR1 pathway is a promising novel therapeutic strategy to treat preeclampsia.


Subject(s)
Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pre-Eclampsia/etiology , Trophoblasts/pathology , Animals , Cell Line , Chemokines/genetics , Female , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Placenta/metabolism , Placenta/pathology , Placenta Growth Factor/metabolism , Pregnancy , Pregnancy Outcome , Trophoblasts/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
3.
Am J Physiol Endocrinol Metab ; 320(4): E786-E796, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33586490

ABSTRACT

We investigated the expression levels of nephroblastoma overexpressed [NOV or CCN3 (cellular communication network factor 3)] in the serum and placenta of pregnant women and of pregnant mice fed a high-fat diet (HFD), and its effect on placental glucose transporter 3 (GLUT3) expression, to examine its role in gestational diabetes mellitus (GDM). NOV/CCN3 expression was increased in the mouse serum during pregnancy. At gestational day 18, NOV/CCN3 protein expression was increased in the serum and placenta of the HFD mice compared with that of mice fed a normal diet. Compared with non-GDM patients, the patients with GDM had significantly increased serum NOV/CCN3 protein expression and placental NOV/CCN3 mRNA expression. Therefore, we hypothesized that NOV/CCN3 signaling may be involved in the pathogenesis of GDM. We administered NOV/CCN3 recombinant protein via intraperitoneal injections to pregnant mice fed HFD or normal diet. NOV/CCN3 overexpression led to glucose intolerance. Combined with the HFD, NOV/CCN3 exacerbated glucose intolerance and caused insulin resistance. NOV/CCN3 upregulates GLUT3 expression and affects the mammalian target of rapamycin (mTOR) pathway in the GDM environment in vivo and in vitro. In summary, our results demonstrate, for the first time, the molecular mechanism of NOV/CCN3 signaling in maternal metabolism to regulate glucose balance during pregnancy. NOV/CCN3 may be a potential target for detecting and treating GDM.NEW & NOTEWORTHY NOV/CCN3 regulates glucose homeostasis in mice during pregnancy. NOV/CCN3 upregulates GLUT3 expression and affects the mTOR pathway in the GDM environment in vivo and in vitro.


Subject(s)
Diet, High-Fat , Glucose Transporter Type 3/genetics , Nephroblastoma Overexpressed Protein/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Dietary Fats/pharmacology , Female , Glucose/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Transporter Type 3/metabolism , Humans , Maternal Nutritional Physiological Phenomena/drug effects , Maternal Nutritional Physiological Phenomena/genetics , Mice , Mice, Inbred C57BL , Nephroblastoma Overexpressed Protein/metabolism , Pregnancy , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
4.
Int J Med Sci ; 18(1): 207-215, 2021.
Article in English | MEDLINE | ID: mdl-33390789

ABSTRACT

Background: Placental-like chondroitin sulfate A (pl-CSA) is exclusively expressed in cancerous and placental tissues and is highly correlated with the degree of malignancy. However, the mechanism through which pl-CSA regulates tumorigenesis and metastasis in choriocarcinoma remains unclear. Methods: Stable transfectants of the JEG3 choriocarcinoma cell line, including a negative control (NC) line and a cell line with knockout of the biosynthetic enzyme CS synthase-2 (ChSy-2) (ChSy-2-/-), were obtained using CRISPR/Cas9 systems and identified by immunofluorescence, flow cytometry, western blots and enzyme-linked immunosorbent assays (ELISAs). The proliferation, migration, invasion and colony formation of the cells were determined by a cell counting kit, scratch-wound assays, transwell assays and soft agar colony formation assays in vitro, respectively. The tumorigenesis and metastasis of choriocarcinoma were also investigated through two xenograft models in vivo. Results: The ChSy-2 protein in the ChSy-2-/-group was below the detection threshold, which was accompanied a significant reduction in the pl-CSA level. Reducing pl-CSA through ChSy-2 knockout significantly inhibited cell proliferation, migration, invasion and colony formation in vitro and tumorigenesis and metastasis of choriocarcinoma, with deceases in tumor volume and metastatic foci and a high percent survival compared to the NC in vivo. Conclusion: pl-CSA, as a necessary component of JEG-3 cells, was efficiently reduced through ChSy-2 knockout, which significantly inhibited the tumorigenesis and metastasis of choriocarcinoma. ChSy-2/pl-CSA could be alternative targets for tumor therapy.


Subject(s)
Carcinogenesis/pathology , Chondroitin Sulfates/metabolism , Choriocarcinoma/secondary , Glycosyltransferases/metabolism , Membrane Proteins/metabolism , Uterine Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Knockdown Techniques , Glycosyltransferases/genetics , Humans , Membrane Proteins/genetics , Mice , Pregnancy , Specific Pathogen-Free Organisms , Xenograft Model Antitumor Assays
5.
Am J Physiol Cell Physiol ; 318(3): C664-C674, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31851527

ABSTRACT

R-spondin3 (RSPO3), an activator of Wnt/ß-catenin signaling, plays a key role in tumorigenesis of various cancers, but its role in choriocarcinoma remains unknown. To investigate the effect of RSPO3 on the tumor growth of choriocarcinoma JEG-3 cells, the expression of RSPO3 in human term placenta was detected, and a stable RSPO3-overexpressing JEG-3 cell line was established via lentivirus-mediated transduction. The expression of biomarkers involved in tumorigenicity was detected in the RSPO3-overexpressing JEG-3 cells, and cell proliferation, invasion, migration, and apoptosis were investigated. Moreover, soft agar clonogenic assays and xenograft tumorigenicity assays were performed to assess the effect of RSPO3 on tumor growth in vitro and in vivo. The results showed that RSPO3 was widely expressed in human term placenta and overexpression of RSPO3 promoted the proliferation and inhibited the migration, invasion, and apoptosis of the JEG-3 cells. Meanwhile, RSPO3 overexpression promoted tumor growth both in vivo and in vitro. Further investigation showed that the phosphorylation levels of Akt, phosphatidylinositol 3-kinase (PI3K), and ERK as well the expression of ß-catenin and proliferating cell nuclear antigen (PCNA) were increased in the RSPO3-overexpressing JEG-3 cells and tumor xenograft. Taken together, these data indicate that RSPO3 promotes the tumor growth of choriocarcinoma via Akt/PI3K/ERK signaling, which supports RSPO3 as an oncogenic driver promoting the progression of choriocarcinoma.


Subject(s)
Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Thrombospondins/biosynthesis , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Adult , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Choriocarcinoma/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pregnancy , Thrombospondins/genetics , Uterine Neoplasms/genetics , Xenograft Model Antitumor Assays/methods
6.
Am J Physiol Endocrinol Metab ; 318(3): E371-E380, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31910029

ABSTRACT

Excess androgen-induced obesity has become a public health problem, and its prevalence has increased substantially in recent years. Chemokine-like receptor 1 (CMKLR1), a receptor of chemerin secreted by adipose tissue, is linked to adipocyte differentiation, adipose tissue development, and obesity. However, the effect of CMKLR1 signaling on androgen-mediated adiposity in vivo remains unclear. Using CMKLR1-knockout mice, we constructed an androgen-excess female mouse model through 5α-dihydrotestosterone (DHT) treatment and an androgen-deficient male mouse model by orchidectomy (ORX). For mechanism investigation, we used 2-(α-Naphthoyl) ethyltrimethylammonium iodide (α-NETA), an antagonist of CMKLR1, to suppress CMKLR1 in vivo and wortmannin, a PI3K signaling antagonist, to treat brown adipose tissue (BAT) explant cultures in vitro. Furthermore, we used histological examination and quantitative PCR, as well as Western blot analysis, glucose tolerance tests, and biochemical analysis of serum, to describe the phenotypes and the changes in gene expression. We demonstrated that excess androgen in the female mice resulted in larger cells in the white adipose tissue (WAT) and the BAT, whereas androgen deprivation in the male mice induced a reduction in cell size. Both of these adipocyte size effects could be attenuated in the CMKLR1-knockout mice. CMKLR1 deficiency influenced the effect of androgen treatment on adipose tissue by regulating the mRNA expression of the androgen receptor (AR) and adipocyte markers (such as Fabp4 and Cidea). Moreover, suppression of CMKLR1 by α-NETA could also reduce the extent of the adipocyte cell enlargement caused by DHT. Furthermore, we found that DHT could reduce the levels of phosphorylated ERK (pERK) in the BAT, while CMKLR1 inactivation inhibited this effect, which had been induced by DHT, through the PI3K signaling pathway. These findings reveal an antiobesity role of CMKLR1 deficiency in regulating lipid accumulation, highlighting the scientific importance for the further development of small-molecule CMKLR1 antagonists as fundamental research tools and/or as potential drugs for use in the treatment of adiposity.


Subject(s)
Androgens/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Receptors, Chemokine/deficiency , Adipocytes/metabolism , Adipocytes/ultrastructure , Adipose Tissue, Brown/drug effects , Androgens/deficiency , Animals , Body Weight , Cell Size , Dihydrotestosterone/pharmacology , Female , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalenes/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/genetics , Wortmannin/pharmacology
7.
Cell Mol Life Sci ; 76(2): 355-367, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30374519

ABSTRACT

The adipokine Chemerin and its receptor, chemokine-like receptor 1 (CMKLR1), are associated with osteoblastogenic differentiation of mesenchymal stem cells (MSCs) and osteoclastogenic differentiation of osteoclast precursors in vitro, suggesting that CMKLR1 would affect the bone mineral density (BMD). However, the role of CMKLR1 on BMD in vivo remains unknown. Here, using CMKLR1 knockout mouse model, we unveiled that CMKLR1 effected the amount of Leydig cells in testis and regulated androgen-dependent bone maintenance in male mice, which exhibited lower serum testosterone levels, thereby reducing the trabecular bone mass. Correspondingly, the mRNA expression of testosterone synthesis enzymes in testis decreased. The bone tissue also showed decreased mRNAs expression of osteogenic markers and increased mRNA levels for osteoclast markers. Furthermore, by in vitro differentiation models, we found CMKLR1-deficiency could break the balance between osteoblastogenesis and osteoclastogenesis that caused a shift from osteogenic to adipogenic differentiation in MSCs and enhanced osteoclast formation. In addition, bone mass increase in CMKLR1 KO male mice can be promoted by treatment with 5α-dihydrotestosterone (DHT), and the inactivation of CMKLR1 in male wild-type (WT) mice with antagonist treatment can lead to low bone mass. Taken together, these data indicate that CMKLR1 positively regulates bone metabolism through mediating testosterone production and the balance between osteoblast and osteoclast formation.


Subject(s)
Bone Density , Receptors, G-Protein-Coupled/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Cell Differentiation , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Femur/diagnostic imaging , Femur/physiology , Interleukin-1beta/analysis , Interleukin-6/analysis , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis , PPAR gamma/genetics , PPAR gamma/metabolism , Receptors, Chemokine , Receptors, G-Protein-Coupled/deficiency , Testis/metabolism , Testis/pathology , Testosterone/biosynthesis , Testosterone/blood , Tibia/diagnostic imaging , Tibia/physiology
8.
Mar Drugs ; 18(9)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32948074

ABSTRACT

Marine microalgae are regarded as potential feedstock because of their multiple valuable compounds, including lipids, pigments, carbohydrates, and proteins. Some of these compounds exhibit attractive bioactivities, such as carotenoids, ω-3 polyunsaturated fatty acids, polysaccharides, and peptides. However, the production cost of bioactive compounds is quite high, due to the low contents in marine microalgae. Comprehensive utilization of marine microalgae for multiple compounds production instead of the sole product can be an efficient way to increase the economic feasibility of bioactive compounds production and improve the production efficiency. This paper discusses the metabolic network of marine microalgal compounds, and indicates their interaction in biosynthesis pathways. Furthermore, potential applications of co-production of multiple compounds under various cultivation conditions by shifting metabolic flux are discussed, and cultivation strategies based on environmental and/or nutrient conditions are proposed to improve the co-production. Moreover, biorefinery techniques for the integral use of microalgal biomass are summarized. These techniques include the co-extraction of multiple bioactive compounds from marine microalgae by conventional methods, super/subcritical fluids, and ionic liquids, as well as direct utilization and biochemical or thermochemical conversion of microalgal residues. Overall, this review sheds light on the potential of the comprehensive utilization of marine microalgae for improving bioeconomy in practical industrial application.


Subject(s)
Biological Products/metabolism , Biotechnology , Microalgae/metabolism , Biological Products/economics , Biological Products/pharmacology , Biomass , Biotechnology/economics , Cost-Benefit Analysis , Energy Metabolism
9.
Am J Physiol Cell Physiol ; 317(3): C556-C565, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31241986

ABSTRACT

Choriocarcinoma is characterized by malignant proliferation and transformation of trophoblasts and is currently treated with systemic chemotherapeutic agents. The lack of specific targets for chemotherapeutic agents results in indiscriminate drug distribution. In our study, we aimed to delineate the mechanism by which G protein-coupled receptor 1 (GPR1) regulates the development of choriocarcinoma and thus investigated GPR1 as a prospective chemotherapeutic target. In this study, GPR1 expression levels were examined in several trophoblast cell lines. We found significantly higher GPR1 expression in choriocarcinoma cells (JEG3 and BeWo) than in normal trophoblast cells (HTR-8/SVneo). Additionally, we studied the role of GPR1 in choriocarcinoma in vitro and in vivo. GPR1 knockdown suppressed proliferation, invasion, and Akt and ERK phosphorylation in vitro and slowed tumor growth in vivo. Interestingly, GPR1 overexpression promoted increased proliferation, invasion, and Akt and ERK phosphorylation in vitro. Furthermore, we identified a specific GPR1-binding seven-amino acid peptide, LRH7-G3, that might also suppress choriocarcinoma in vitro and in vivo through phage display. Our study is the first to report that GPR1 may play a role in regulating choriocarcinoma progression through the Akt and ERK pathways. GPR1 could be a promising potential pharmaceutical target for choriocarcinoma.


Subject(s)
Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Receptors, G-Protein-Coupled/physiology , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Invasiveness/pathology , Pregnancy
10.
Am J Physiol Endocrinol Metab ; 316(6): E987-E997, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30835511

ABSTRACT

Chemerin and G protein-coupled receptor 1 (GPR1) are increased in serum and placenta in mice during pregnancy. Interestingly, we observed increased serum chemerin levels and decreased GPR1 expression in placenta of high-fat-diet-fed mice compared with chow-fed mice at gestational day 18. GPR1 protein and gene levels were significantly decreased in gestational diabetes mellitus (GDM) patient placentas. Therefore, we hypothesized that chemerin/GPR1 signaling might participate in the pathogenic mechanism of GDM. We investigated the role of GPR1 in carbohydrate homeostasis during pregnancy using pregnant mice transfected with small interfering RNA for GPR1 or a negative control. GPR1 knockdown exacerbated glucose intolerance, disrupted lipid metabolism, and decreased ß-cell proliferation and insulin levels. Glucose transport protein-3 and fatty acid binding protein-4 were downregulated with reducing GPR1 in vivo and in vitro via phosphorylated AKT pathway. Taken together, our findings first demonstrate the expression of GPR1, the characterization of its direct biological effects in humans and mice, as well as the molecular mechanism that indicates the role of GPR1 signaling in maternal metabolism during pregnancy, suggesting a novel feedback mechanism to regulate glucose balance during pregnancy, and GPR1 could be a potential target for the detection and therapy of GDM.


Subject(s)
Chemokines/genetics , Diabetes, Gestational/genetics , Pregnancy/metabolism , Receptors, G-Protein-Coupled/genetics , Adult , Animals , Blood Glucose/metabolism , Cell Proliferation/genetics , Chemokines/metabolism , Diabetes, Gestational/metabolism , Diet, High-Fat , Fatty Acid-Binding Proteins/metabolism , Female , Gene Knockdown Techniques , Glucose Transporter Type 3/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Intercellular Signaling Peptides and Proteins , Lipid Metabolism/genetics , Mice , Placenta , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
Int J Mol Sci ; 20(21)2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31683965

ABSTRACT

The available and effective therapeutic means to treat choriocarcinoma is seriously lacking, mainly due to the toxic effects caused by chemotherapy and radiotherapy. Accordingly, we developed a method for targeting delivery of chemotherapeutical drugs only to cancer cells, not normal cells, in vivo, by using a synthetic placental chondroitin sulfate (CSA)-binding peptide (plCSA-BP) derived from malarial protein VAR2CSA. A 28 amino acids placental CSA-binding peptide (plCSA-BP) from the VAR2CSA was synthesized as a guiding peptide for tumor-targeting delivery, dendrigraft poly-L-lysines (DGL) was modified with plCSA-BP and served as a novel targeted delivery carrier. Choriocarcinoma was selected to test the effect of targeted delivery carrier, and prodigiosin isolated from Serratia marcescens subsp. lawsoniana was selected as a chemotherapeutical drug and encapsulated in the DGL modified by the plCSA-BP nanoparticles (DGL/CSA-PNPs). DGL/CSA-PNPs had a sustained slow-release feature at pH 7.4, which could specifically bind to the JEG3 cells and exhibited better anticancer activity than that of the controls. The DGL/CSA-PNPs induced the apoptosis of JEG3 cells through caspase-3 and the P53 signaling pathway. DGL/CSA-PNPs can be used as an excellent targeted delivery carrier for anticancer drugs, and the prodigiosin could be an alternative chemotherapeutical drug for choriocarcinoma.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Choriocarcinoma/pathology , Nanoparticles/chemistry , Peptides/chemistry , Polylysine/chemistry , Prodigiosin/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Chondroitin Sulfates/chemistry , Choriocarcinoma/metabolism , Drug Compounding , Drug Delivery Systems/methods , Drug Liberation , Humans , Prodigiosin/administration & dosage , Prodigiosin/chemistry , Reproducibility of Results
12.
Reprod Biol Endocrinol ; 16(1): 50, 2018 May 24.
Article in English | MEDLINE | ID: mdl-29793502

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a complex genetic disease with multifarious phenotypes. Many researches use dehydroepiandrosterone (DHEA) to induce PCOS in pubertal mouse models. The aim of this study was to investigate the role of GPR1 in dehydroepiandrosterone (DHEA)-induced hyperandrogenized mice. METHODS: Prepubertal C57BL/6 mice (25 days of age) and Gpr1-deficient mice were each divided into two groups and injected daily with sesame oil with or without DHEA (6 mg/100 g) for 21 consecutive days. Hematoxylin and eosin (H&E) staining was performed to determine the characteristics of the DHEA-treated ovaries. Real-time PCR was used to examine steroid synthesis enzymes gene expression. Granulosa cell was cultured to explore the mechanism of DHEA-induced, GPR1-mediated estradiol secretion. RESULTS: DHEA treatment induced some aspects of PCOS in wild-type mice, such as increased body weight, elevated serum testosterone, increased number of small, cystic, atretic follicles, and absence of corpus luteum in ovaries. However, Gpr1 deficiency significantly attenuated the DHEA-induced weight gain and ovarian phenotype, improving steroidogenesis in ovaries and estradiol synthesis in cultured granulosa cells, partially through mTOR signaling. CONCLUSIONS: In conclusion, Gpr1 deficiency leads to the improvement of steroid synthesis in mice hyperandrogenized with DHEA, indicating that GPR1 may be a therapeutic target for DHEA-induced hyperandrogenism.


Subject(s)
Hyperandrogenism/blood , Hyperandrogenism/genetics , Receptors, G-Protein-Coupled/genetics , Testosterone/blood , Animals , Cells, Cultured , Dehydroepiandrosterone , Disease Models, Animal , Estradiol/blood , Female , Hyperandrogenism/chemically induced , Hyperandrogenism/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology
13.
Bioprocess Biosyst Eng ; 40(7): 1091-1100, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28455664

ABSTRACT

The ratio of carbon to nitrogen (C/N) in media plays a crucial role in the production of microbial carotenoids. However, the effects of a high C/N ratio on carotenoid production are ambiguous, and the mechanism of how C/N ratio affects astaxanthin accumulation in X. dendrorhous is unclear. In this study, the influence of C/N ratio on astaxanthin biosynthesis in X. dendrorhous at a fixed nitrogen concentration was investigated, and comparative proteomics were applied to address how C/N ratio affects cell growth and astaxanthin accumulation in X. dendrorhous. The results showed that cell growth and astaxanthin accumulation in X. dendrorhous were strongly related to the ratio of carbon to nitrogen with increasing C/N ratio in medium. However, the astaxanthin content per cell showed an inverse relationship, decreasing with an increasing C/N ratio. Differential proteomics showed the proteins with highest degree of change in expression under varying C/N ratios were mainly involved in carbohydrate metabolic pathways and carotenogenesis metabolism. In addition, several redox- and stress-associated proteins were up-regulated along with the carotenogenesis proteins, implying the environmental stress may affect metabolism and astaxanthin synthesis. A possible regulatory mechanism in response to glucose in X. dendrorhous is discussed.


Subject(s)
Basidiomycota , Carbon , Proteomics , Xanthophylls
14.
Plant J ; 81(1): 95-107, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25353310

ABSTRACT

Astaxanthin, a red ketocarotenoid with strong antioxidant activity and high commercial value, possesses important physiological functions in astaxanthin-producing microalgae. The green microalga Haematococcus pluvialis accumulates up to 4% fatty acid-esterified astaxanthin (by dry weight), and is used as a model species for exploring astaxanthin biosynthesis in unicellular photosynthetic organisms. Although coordination of astaxanthin and fatty acid biosynthesis in a stoichiometric fashion was observed in H. pluvialis, the interaction mechanism is unclear. Here we dissected the molecular mechanism underlying coordination between the two pathways in H. pluvialis. Our results eliminated possible coordination of this inter-dependence at the transcriptional level, and showed that this interaction was feedback-coordinated at the metabolite level. In vivo and in vitro experiments indicated that astaxanthin esterification drove the formation and accumulation of astaxanthin. We further showed that both free astaxanthin biosynthesis and esterification occurred in the endoplasmic reticulum, and that certain diacylglycerol acyltransferases may be the candidate enzymes catalyzing astaxanthin esterification. A model of astaxanthin biosynthesis in H. pluvialis was subsequently proposed. These findings provide further insights into astaxanthin biosynthesis in H. pluvialis.


Subject(s)
Chlorophyta/metabolism , Fatty Acids/biosynthesis , Microalgae/metabolism , Algal Proteins/metabolism , Algal Proteins/physiology , Chlorophyta/genetics , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/physiology , Endoplasmic Reticulum/metabolism , Esterification , Metabolic Networks and Pathways , Transcription, Genetic , Xanthophylls/biosynthesis
15.
Biotechnol Bioeng ; 113(10): 2088-99, 2016 10.
Article in English | MEDLINE | ID: mdl-27563850

ABSTRACT

The unicellular green alga Haematococcus pluvialis has been exploited as a cell factory to produce the high-value antioxidant astaxanthin for over two decades, due to its superior ability to synthesize astaxanthin under adverse culture conditions. However, slow vegetative growth under favorable culture conditions and cell deterioration or death under stress conditions (e.g., high light, nitrogen starvation) has limited the astaxanthin production. In this study, a new paradigm that integrated heterotrophic cultivation, acclimation of heterotrophically grown cells to specific light/nutrient regimes, followed by induction of astaxanthin accumulation under photoautotrophic conditions was developed. First, the environmental conditions such as pH, carbon source, nitrogen regime, and light intensity, were optimized to induce astaxanthin accumulation in the dark-grown cells. Although moderate astaxanthin content (e.g., 1% of dry weight) and astaxanthin productivity (2.5 mg L(-1) day(-1) ) were obtained under the optimized conditions, a considerable number of cells died off when subjected to stress for astaxanthin induction. To minimize the susceptibility of dark-grown cells to light stress, the algal cells were acclimated, prior to light induction of astaxanthin biosynthesis, under moderate illumination in the presence of nitrogen. Introduction of this strategy significantly reduced the cell mortality rate under high-light and resulted in increased cellular astaxanthin content and astaxanthin productivity. The productivity of astaxanthin was further improved to 10.5 mg L(-1) day(-1) by implementation of such a strategy in a bubbling column photobioreactor. Biochemical and physiological analyses suggested that rebuilding of photosynthetic apparatus including D1 protein and PsbO, and recovery of PSII activities, are essential for acclimation of dark-grown cells under photo-induction conditions. Biotechnol. Bioeng. 2016;113: 2088-2099. © 2016 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals, Inc.


Subject(s)
Chlorophyta/metabolism , Nitrogen/metabolism , Photobioreactors/microbiology , Photosynthesis/physiology , Cell Culture Techniques/methods , Dose-Response Relationship, Radiation , Light , Photic Stimulation/methods , Photosynthesis/radiation effects , Radiation Dosage , Xanthophylls/biosynthesis , Xanthophylls/isolation & purification , Xanthophylls/radiation effects
16.
J Exp Bot ; 65(15): 4317-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821952

ABSTRACT

Haematococcus pluvialis cells predominantly remain in the macrozooid stage under favourable environmental conditions but are rapidly differentiated into haematocysts upon exposure to various environmental stresses. Haematocysts are characterized by massive accumulations of astaxanthin sequestered in cytosolic oil globules. Lipidomic analyses revealed that synthesis of the storage lipid triacylglycerol (TAG) was substantially stimulated under high irradiance. Simultaneously, remodelling of membrane glycerolipids occurred as a result of dramatic reductions in chloroplast membrane glycolipids but remained unchanged or declined slightly in extraplastidic membrane glycerolipids. De novo assembly of transcriptomes revealed the genomic and metabolic features of this unsequenced microalga. Comparative transcriptomic analysis showed that so-called resting cells (haematocysts) may be more active than fast-growing vegetative cells (macrozooids) regarding metabolic pathways and functions. Comparative transcriptomic analyses of astaxanthin biosynthesis suggested that the non-mevalonate pathway mediated the synthesis of isopentenyl diphosphate, as the majority of genes involved in subsequent astaxanthin biosynthesis were substantially up-regulated under high irradiance, with the genes encoding phytoene synthase, phytoene desaturase, and ß-carotene hydroxylase identified as the most prominent regulatory components. Accumulation of TAG under high irradiance was attributed to moderate up-regulation of de novo fatty acid biosynthesis at the gene level as well as to moderate elevation of the TAG assembly pathways. Additionally, inferred from transcriptomic differentiation, an increase in reactive oxygen species (ROS) scavenging activity, a decrease in ROS production, and the relaxation of over-reduction of the photosynthetic electron transport chain will work together to protect against photooxidative stress in H. pluvialis under high irradiance.


Subject(s)
Chlorophyta/metabolism , Lipid Metabolism , Oxidative Stress , Transcriptome , Chlorophyta/radiation effects , Molecular Sequence Annotation , Sequence Analysis, DNA , Sunlight , Xanthophylls/biosynthesis
17.
Food Chem ; 447: 138873, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38452536

ABSTRACT

Food-derived angiotensin-converting enzyme-inhibitory (ACE-I) peptides have attracted extensive attention. Herein, the ACE-I peptides from Scomber japonicus muscle hydrolysates were screened, and their mechanisms of action and inhibition stability were explored. The quantitative structure-activity relationship (QSAR) model based on 5z-scale metrics was developed to rapidly screen for ACE-I peptides. Two novel potential ACE-I peptides (LTPFT, PLITT) were predicted through this model coupled with in silico screening, of which PLITT had the highest activity (IC50: 48.73 ± 7.59 µM). PLITT inhibited ACE activity with a mixture of non-competitive and competitive mechanisms, and this inhibition mainly contributed to the hydrogen bonding based on molecular docking study. PLITT is stable under high temperatures, pH, glucose, and NaCl. The zinc ions (Zn2+) and copper ions (Cu2+) enhanced ACE-I activity. The study suggests that the QSAR model is effective in rapidly screening for ACE-I inhibitors, and PLITT can be supplemented in foods to lower blood pressure.


Subject(s)
Protein Hydrolysates , Quantitative Structure-Activity Relationship , Molecular Docking Simulation , Protein Hydrolysates/pharmacology , Protein Hydrolysates/chemistry , Peptides/pharmacology , Peptides/chemistry , Muscles/metabolism , Ions , Angiotensins , Peptidyl-Dipeptidase A/metabolism
18.
Bioresour Bioprocess ; 10(1): 29, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-38647925

ABSTRACT

Astaxanthin is an important ketocarotenoid widely used in industries. However, its application is limited because of its low yield. Sodium citrate (Na-citrate), one of the major carbon sources for microorganisms, can promote cell growth and product accumulation. The basidiomycetous red yeast Xanthophyllomyces dendrorhous was thus used to study the effect of Na-citrate on cell growth and astaxanthin synthesis. The highest biomass and astaxanthin yield (6.0 g/L and 22.5 mg/L) were obtained in shake-flask when 3 g/L Na-citrate was added at 24 h and were 1.8 and 2.0 times higher than those of the control group, respectively. Furthermore, metabolomics and real-time reverse transcription PCR (qRT-PCR) analysis were conducted to study the metabolic pathways of X. dendrorhous in response to Na-citrate. The qRT-PCR assay revealed that Na-citrate facilitated glucose consumption, promoted the metabolic flux from glycolysis, and regulated the tricarboxylic acid (TCA) cycle, providing more energy and substrates for the synthesis of astaxanthin. The gene analysis revealed that adding Na-citrate significantly upregulated the expression of six key genes (ICL, HMGS, crtE, crtYB, crtI, and crtS) involved in pathways related to astaxanthin biosynthesis. These results suggest that exogenous Na-citrate treatment is a potentially valuable strategy to stimulate astaxanthin production in X. dendrorhous.

19.
Bioresour Technol ; 362: 127761, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35961507

ABSTRACT

Mixotrophy of Haematococcus pluvialis is a potential strategy for producing astaxanthin. However, this strategy has not been extensively commercialized because the mixotrophic mechanisms by which H. pluvialis overcomes high light stress are unclear. This study analyzed the biochemical compositions and differential proteomics of mixotrophic H. pluvialis under different light conditions. High light exposure substantially increased astaxanthin, carbohydrate, and fatty acid contents. A total of 119 and 81 proteins were significantly up- and down-regulated after two days of high light exposure. These proteins mainly enriched pathways for photosynthetic metabolism, glyoxylate cycle, and biosynthesis of secondary metabolites. This study proposed a regulatory model through which mixotrophic H. pluvialis copes with high light stress. The model includes pathways for modulating photosynthetic apparatus, increasing astaxanthin accumulation by enhancing photorespiration, pentose phosphate and Embden-Meyerhof-Parna pathways, while thickening the cell wall by malate-oxaloacetate shuttle.


Subject(s)
Carbon , Chlorophyceae , Fatty Acids , Light , Photosynthesis
20.
Foods ; 11(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36010470

ABSTRACT

Oxyresveratrol (Oxy) has attracted much attention by employing it as an antibrowning agent in fruits and vegetables. In this study, the formation of cyclodextrin (CD) inclusion exhibited a certain protective effect on Oxy oxidative degradation, while hydroxypropyl-ß-cyclodextrin (HP-ß-CD) inclusion complex showed stronger stabilizing effects than those of ß-cyclodextrin (ß-CD). The combined use of CD and hydroxypropyl methylcellulose (HPMC) greatly improved the stability of Oxy-CD inclusion complexes, with approximately 70% of the trans-Oxy retained after 30 days of storage under light conditions at 25 °C. The results of the interaction between CD and Oxy determined by phase solubility studies and fluorescence spectroscopic analysis showed that the binding strength of CD and Oxy increased in the presence of HPMC. Moreover, Oxy combined with ascorbic acid and HPMC showed an excellent antibrowning effect on fresh-cut apple slices during the 48 h test period, indicating that adding HPMC as the third component will not influence the antibrowning activity of Oxy.

SELECTION OF CITATIONS
SEARCH DETAIL