Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 651
Filter
Add more filters

Publication year range
1.
Trends Immunol ; 43(1): 41-50, 2022 01.
Article in English | MEDLINE | ID: mdl-34844850

ABSTRACT

Catestatin (CST) is a bioactive cleavage product of the neuroendocrine prohormone chromogranin A (CgA). Recent findings show that CST can exert anti-inflammatory and antiadrenergic effects by suppressing the inflammatory actions of mammalian macrophages. However, recent findings also suggest that macrophages themselves are major CST producers. Here, we hypothesize that macrophages produce CST in an inflammation-dependent manner and thereby might self-regulate inflammation in an autocrine fashion. CST is associated with pathological conditions hallmarked by chronic inflammation, including autoimmune, cardiovascular, and metabolic disorders. Since intraperitoneal injection of CST in mouse models of diabetes and inflammatory bowel disease has been reported to be beneficial for mitigating disease, we posit that CST should be further investigated as a candidate target for treating certain inflammatory diseases.


Subject(s)
Inflammation , Peptide Fragments , Animals , Chromogranin A/metabolism , Humans , Macrophages , Mammals , Mice
2.
Eur Heart J ; 44(19): 1732-1744, 2023 05 14.
Article in English | MEDLINE | ID: mdl-36861348

ABSTRACT

AIMS: Members of the chromogranin family play a role in angiogenesis. One such biologically active peptide, generated through the processing of chromogranin A, is vasostatin-2. This study aimed at assessing the association of serum vasostatin-2 levels with coronary collateral vessels (CCV) in diabetic patients with chronic total occlusions (CTO) and the effects of vasostatin-2 on angiogenesis in diabetic mice with hindlimb or myocardial ischemia. METHODS AND RESULTS: Serum levels of vasostatin-2 in 452 diabetic CTO patients were evaluated. The status of CCV was categorized according to the Rentrop score. Vasostatin-2 recombinant protein or phosphate-buffered saline were then injected intraperitoneally in diabetic mouse models of hindlimb or myocardial ischemia, followed by laser Doppler imaging and molecular biology examinations. The effects of vasostatin-2 were also ascertained in endothelial cells and macrophages, with mechanisms clarified using ribonucleic acid (RNA) sequencing. Serum levels of vasostatin-2 were significantly different and progressively higher across Rentrop score 0, 1, 2, and 3 groups (P < .001), with significantly lower levels in patients with poor CCV (Rentrop score 0 and 1) than in those with good CCV (Rentrop score 2 and 3) (P < .05). Vasostatin-2 significantly promoted angiogenesis in diabetic mice with hindlimb or myocardial ischemia. RNA-seq analyzes verified an angiotensin-converting enzyme 2 (ACE2)-mediated vasostatin-2-induction of angiogenesis in ischemic tissues. CONCLUSION: Lower serum levels of vasostatin-2 are associated with poor CCV in diabetic CTO patients compared with patients with good CCV. Vasostatin-2 significantly promotes angiogenesis in diabetic mice with hindlimb or myocardial ischemia. Such effects are mediated by ACE2.


Subject(s)
Coronary Artery Disease , Diabetes Mellitus, Experimental , Myocardial Ischemia , Mice , Animals , Chromogranin A/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Endothelial Cells/metabolism , Diabetes Mellitus, Experimental/metabolism , Coronary Artery Disease/metabolism , Collateral Circulation
3.
Int J Mol Sci ; 25(5)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38473713

ABSTRACT

Preeclampsia (PE) is a unique pregnancy disorder affecting women across the world. It is characterized by the new onset of hypertension with coexisting end-organ damage. Although the disease has been known for centuries, its exact pathophysiology and, most importantly, its prevention remain elusive. The basis of its associated molecular changes has been attributed to the placenta and the hormones regulating its function. One such hormone is chromogranin A (CgA). In the placenta, CgA is cleaved to form a variety of biologically active peptides, including catestatin (CST), known inter alia for its vasodilatory effects. Recent studies indicate that the CST protein level is diminished both in patients with hypertension and those with PE. Therefore, the aim of the present paper is to review the most recent and most relevant in vitro, in vivo, and clinical studies to provide an overview of the proposed impact of CST on the molecular processes of PE and to consider the possibilities for future experiments in this area.


Subject(s)
Hypertension , Pre-Eclampsia , Humans , Female , Chromogranin A/metabolism , Peptide Fragments/metabolism
4.
Amino Acids ; 55(5): 549-561, 2023 May.
Article in English | MEDLINE | ID: mdl-36914766

ABSTRACT

Chromogranin A (ChgA) is an acidic pro-protein found in neuroendocrine organs, pheochromocytoma chromaffin granules, and tumor cells. Proteolytic processing of ChgA gives rise to an array of biologically active peptides such as pancreastatin (PST), vasostatin, WE14, catestatin (CST), and serpinin, which have diverse roles in regulating cardiovascular functions and metabolism, as well as inflammation. Intricate tissue-specific role of ChgA-derived peptide activity in preclinical rodent models of metabolic syndrome reveals complex effects on carbohydrate and lipid metabolism. Indeed, ChgA-derived peptides, PST and CST, play a pivotal role in metabolic syndrome such as obesity, insulin resistance, and diabetes mellitus. Additionally, supplementation of specific peptide in ChgA-KO mice have an opposing effect on physiological functions, such as PST supplementation reduces insulin sensitivity and enhances inflammatory response. In contrast, CST supplementation enhances insulin sensitivity and reduces inflammatory response. In this review, we focus on the tissue-specific role of PST and CST as therapeutic targets in regulating carbohydrate and lipid metabolism, along with the associated risk factors.


Subject(s)
Diabetes Mellitus , Insulin Resistance , Metabolic Syndrome , Mice , Animals , Chromogranin A/pharmacology , Chromogranin A/metabolism , Metabolic Syndrome/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Peptides , Diabetes Mellitus/drug therapy , Carbohydrates
5.
Neuroendocrinology ; 113(5): 501-518, 2023.
Article in English | MEDLINE | ID: mdl-36473454

ABSTRACT

INTRODUCTION: Cancer-associated fibroblasts (CAF) have been identified as relevant contributors to cancer progression and drug resistance in many tumors. Although neuroendocrine tumors (NET) are often associated with a strong stromal reaction, no study has addressed whether CAF are involved in progression and therapeutic resistance in NET. The aim of this study was to characterize the role of CAF in NET. METHODS: We established primary CAF cultures derived from NET liver metastases to study the effect on NET cell lines NT-3 and BON. Immunohistochemistry was performed on tissue sections of primary and metastatic NET tissue. RESULTS: Immunohistochemistry identified CAF dispersed in between tumor cells and within fibrotic bands separating tumor cell clusters in NET. Stimulating NET cells with CAF decreased expression of SSTR2 and chromogranin A and induced expression of CXCR4. CAF induced a 2.3-fold increase in proliferation and completely reversed the response to everolimus in NT-3 cells. We identified STAT3 as the main signaling pathway induced by CAF. STAT3 targeting by small interfering RNA knockdown and inhibitors prevented CAF-induced proliferation and restored everolimus responsiveness. STAT3 activation in NET tissue was associated with decreased chromogranin A expression, increased Ki-67 index, and decreased 5-year overall and progression-free survival. CAF directly influence proliferation and therapeutic response in NET cells. CONCLUSION: Identifying STAT3 as the contributing pathway of this so far neglected tumor-stroma interaction has the potential to become a new therapeutic target to halt tumor growth and to restore therapeutic responsiveness in NET.


Subject(s)
Cancer-Associated Fibroblasts , Neuroendocrine Tumors , Humans , Everolimus/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Neuroendocrine Tumors/pathology , Drug Resistance, Neoplasm , Chromogranin A/metabolism , Cell Line, Tumor , Cell Proliferation , STAT3 Transcription Factor/metabolism
6.
Int J Mol Sci ; 24(5)2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36902417

ABSTRACT

Pregnancy is a state of physiological and hormonal changes. One of the endocrine factors involved in these processes is chromogranin A, an acidic protein produced, among others, by the placenta. Although it has been previously linked to pregnancy, no existing articles have ever managed to clarify the role of this protein regarding this subject. Therefore, the aim of the present study is to gather knowledge of chromogranin A's function with reference to gestation and parturition, clarify elusive information, and, most importantly, to formulate hypotheses for the future studies to verify.


Subject(s)
Chromogranins , Endocrine System , Pregnancy , Female , Humans , Chromogranin A/metabolism , Chromogranins/metabolism , Endocrine System/metabolism , Parturition , Placenta/metabolism , Peptide Fragments/metabolism
7.
Biochem Biophys Res Commun ; 605: 162-170, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35339012

ABSTRACT

CGA47-66 (Chromofungin, CHR), is a peptide derived from the N-terminus of chromogranin A (CgA), has been proven to inhibit the lipopolysaccharide (LPS)-induced brain injury. However, the underlying mechanism is still unknown. We found that CGA47-66 exerted a protective effect on cognitive impairment by inhibiting the destruction of the blood-brain barrier (BBB) in the LPS-induced sepsis mice model. In addition, the hCMEC/D3 cell line was used to establish an in vitro BBB model. Under LPS stimulation, CGA47-66 could significantly alleviate the hyperpermeability of the BBB, the destruction of tight junction proteins, and the rearrangement of F-actin. To investigate the underlying mechanism, we used LY294002, a PI3K inhibitor, which partially reduced the protective effect of CGA47-66 on the integrity of BBB. Indicating that the PI3K/AKT pathway plays a vital role in the brain-protective function of CGA47-66, which might be a potential therapeutic target for septic brain injury.


Subject(s)
Blood-Brain Barrier , Brain Injuries , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/prevention & control , Chromogranin A/metabolism , Chromogranin A/pharmacology , Lipopolysaccharides/pharmacology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
8.
Biochem J ; 478(10): 1907-1919, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33955460

ABSTRACT

Antimicrobial peptides (AMPs) are conventional antibiotic alternatives due to their broad-spectrum antimicrobial activities and special mechanisms of action against pathogens. The antifungal peptide CGA-N12 was originally derived from human chromogranin A (CGA) and consists of the 65th to 76th amino acids of the CGA N-terminal region. In the present study, we found that CGA-N12 had fungicidal activity and exhibited time-dependent inhibition activity against Candida tropicalis. CGA-N12 entered the cells to exert its antagonist activity. The internalization of CGA-N12 was energy-dependent and accompanied by actin cytoskeleton-, clathrin-, sulfate proteoglycan-, endosome-, and lipid-depleting agent-mediated endocytosis. Moreover, the CGA-N12 internalization pathway was related to the peptide concentration. The effects of CGA-N12 on the cell membrane were investigated. CGA-N12 at a low concentration less than 4 × MIC100 did not destroy the cell membrane. While with increasing concentration, the damage to the cell membrane caused by CGA-N12 became more serious. At concentrations greater than 4 × MIC100, CGA-N12 destroyed the cell membrane integrity. Therefore, the membrane activity of CGA-N12 is concentration dependant.


Subject(s)
Antifungal Agents/pharmacology , Candida tropicalis/drug effects , Cell Membrane/metabolism , Chromogranin A/metabolism , Endocytosis , Pore Forming Cytotoxic Proteins/pharmacology , Biological Transport , Calcium/metabolism , Candida tropicalis/growth & development , Cell Membrane/drug effects , Humans
9.
Int J Mol Sci ; 23(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36362289

ABSTRACT

Little is known about the adaptor protein FAM159B. Recently, FAM159B was shown to be particularly expressed in neuroendocrine cells and tissues, such as pancreatic islets and neuroendocrine cells of the bronchopulmonary and gastrointestinal tracts, as well as in different types of neuroendocrine tumours. To gain insights into possible interactions of FAM159B with other proteins and/or receptors, we analysed the co-expression of FAM159B and various neuroendocrine-specific markers in the cancer cell lines BON-1, PC-3, NCI-h82, OH-1, and A431 and also in human pancreatic tissues and pancreatic neuroendocrine tumours. The markers included prominent markers of neuroendocrine differentiation, such as chromogranin A (CgA), neuron-specific enolase (NSE), synaptophysin (SYP), insulinoma-associated protein 1 (INSM1), neural cell adhesion molecule 1 (NCAM1), serotonin (5-HT), somatostatin-14/28 (SST), and several receptors that are typically expressed by neuroendocrine cells, such as dopamine receptor 2 (D2R), somatostatin receptor (SSTR) 1, 2, 3, 4 and 5, and regulator of G-protein signalling 9 (RGS9). FAM159B was expressed evenly throughout the cytosol in all five cancer cell lines. Immunocytochemical and immunohistochemical analyses revealed co-expression of FAM159B with SYP, INSM1, RGS9, D2R, SSTR2, SSTR3, SSTR4, and SSTR5 and strong overlapping co-localisation with NSE. Double-labelling and co-immunoprecipitation Western blot analyses confirmed a direct association between FAM159B and NSE. These results suggest the involvement of FAM159B in several intracellular signalling pathways and a direct or indirect influence on diverse membrane proteins and receptors.


Subject(s)
Neuroendocrine Cells , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Neuroendocrine Cells/metabolism , Biomarkers, Tumor/metabolism , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Chromogranin A/genetics , Chromogranin A/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Pancreatic Neoplasms/pathology , Repressor Proteins/metabolism
10.
Molecules ; 27(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35163861

ABSTRACT

The aim of this study is to reveal the potential roles of apoptosis markers (Bcl2 and p53), proliferation markers (Ki-67 and CyclD1), and the neuroendocrine marker Chromogranin A as markers for the radioresistance of rectal cancer. Statistically significant differences were found in the expression of p53, Ki-67, and Chromogranin A in groups of patients with and without a favorable prognosis after radiotherapy. The survival analysis revealed that the marker of neuroendocrine differentiation, Chromogranin A, also demonstrated a high prognostic significance, indicating a poor prognosis. Markers of proliferation and apoptosis had no prognostic value for patients who received preoperative radiotherapy. Higher Chromogranin A values were predictors of poor prognosis. The results obtained from studying the Chromogranin A expression suggest that the secretion of biologically active substances by neuroendocrine cells causes an increase in tumor aggressiveness.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Immunohistochemistry/methods , Neuroendocrine Cells/pathology , Rectal Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/radiotherapy , Adult , Aged , Chromogranin A/metabolism , Cyclin D1/metabolism , Female , Follow-Up Studies , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Neuroendocrine Cells/metabolism , Prognosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Rectal Neoplasms/metabolism , Rectal Neoplasms/radiotherapy , Survival Rate , Tumor Suppressor Protein p53/metabolism
11.
J Cell Physiol ; 236(11): 7745-7758, 2021 11.
Article in English | MEDLINE | ID: mdl-34061983

ABSTRACT

The biosynthesis of many of the peptides involved in homeostatic control requires peptidylglycine α-amidating monooxygenase (PAM), an ancient, highly conserved copper- and ascorbate-dependent enzyme. Using the production of amidated chromogranin A to monitor PAM function in tumor cells, physiologically relevant levels of hypoxia were shown to inhibit this monooxygenase. The ability of primary pituitary cells exposed to hypoxic conditions for 4 h to produce amidated chromogranin A was similarly inhibited. The affinity of the purified monooxygenase for oxygen (Km = 99 ± 19 µM) was consistent with this result. The ability of PAM to alter secretory pathway behavior under normoxic conditions required its monooxygenase activity. Under normoxic conditions, hypoxia-inducible factor 1a levels in dense cultures of corticotrope tumor cells expressing high levels of PAM exceeded those in control cells; expression of inactive monooxygenase did not have this effect. The effects of hypoxia on levels of two PAM-regulated genes (activating transcription factor 3 [Atf3] and FK506 binding protein 2 [Fkbp2]) differed in cells expressing high versus low levels of PAM. Putative hypoxia response elements occur in both human and mouse PAM, and hPAM has consistently been identified as one of the genes upregulated in response to hypoxia. Expression of PAM is also known to alter gene expression. A quarter of the genes consistently upregulated in response to hypoxia were downregulated following increased expression of PAM. Taken together, our data suggest roles for PAM and amidated peptide secretion in the coordination of tissue-specific responses to hypoxia.


Subject(s)
Chromogranin A/metabolism , Mixed Function Oxygenases/metabolism , Multienzyme Complexes/metabolism , Pituitary Gland, Anterior/enzymology , Pituitary Neoplasms/enzymology , Tumor Hypoxia , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Amidine-Lyases/genetics , Amidine-Lyases/metabolism , Animals , Cell Line, Tumor , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mixed Function Oxygenases/genetics , Multienzyme Complexes/genetics , Pituitary Gland, Anterior/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Rats , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
12.
Prostate ; 81(9): 508-520, 2021 06.
Article in English | MEDLINE | ID: mdl-33955576

ABSTRACT

PURPOSE: Hedgehog (Hh) signaling promotes castration-resistant prostate cancer by supporting androgen-independent prostate cancer cell development and growth; however, its role in neuroendocrine prostate cancer (NEPC) has not yet been explored. In this study, we assessed the expression of key genes involved in Hh signaling in prostate cancer and investigated the potential role of smoothened (SMO) in the pathogenesis of NEPC. METHODS: Six public datasets, each containing cases of prostate adenocarcinoma (AdPC) and NEPC, were analyzed to compare the differential messenger RNA (mRNA) expression of six classic Hh signaling genes. The SMO, synaptophysin, chromogranin A (CHGA) and androgen receptor (AR) proteins were evaluated in human tissues from 5 cases of NEPC, 2 cases of AdPC mixed with NEPC, 2 cases of AdPC with neuroendocrine differentiation and 22 cases of high-grade AdPC as determined by an immunohistochemistry assay. Gene set enrichment analysis (GSEA) was performed to identify relevant genetic signatures associated with SMO expression based on the public datasets. Stable SMO-knockdown LNCaP and C4-2B cells were established with a lentiviral system, and the expression of SMO, Gli1, AR, prostate-specific antigen (PSA), and REST was assessed by real-time polymerase chain reaction and western blot. Secreted PSA in the conditioned medium was assessed by ELISA. Gli1 was ectopically expressed performed by the transfection of Gli1 complementary DNA into SMO-knockdown LNCaP cells, and western blot was used to assess of AR and PSA expression. RESULTS: The mRNA level of SMO was dramatically downregulated in NEPC samples compared with AdPC samples in all 6 public datasets. SMO protein loss was observed in 100% of NEPC samples but in only 9% (2 of 22) of high-grade AdPC samples. GSEA results showed that SMO loss was closely correlated with AR signaling activity. Stable SMO knockdown significantly attenuated AR signaling activity and suppressed AR expression, while Gli1 overexpression partially reversed the inhibitory effects of SMO knockdown on AR signaling activity and AR expression in LNCaP and C4-2B cells. CONCLUSION: These results demonstrate that SMO loss is a characteristic of NEPC and that detecting SMO by IHC could aid pathologists in NEPC diagnosis. SMO loss may promote NEPC pathogenesis by modulating AR signaling.


Subject(s)
Adenocarcinoma , Carcinoma, Neuroendocrine , Prostate , Prostatic Neoplasms , Smoothened Receptor , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cells, Cultured , Chromogranin A/metabolism , Down-Regulation , Gene Expression Profiling , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Humans , Immunohistochemistry , Male , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/analysis , Receptors, Androgen/metabolism , Signal Transduction/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Synaptophysin/metabolism
13.
FASEB J ; 34(5): 6769-6790, 2020 05.
Article in English | MEDLINE | ID: mdl-32227388

ABSTRACT

Chromogranin A (CgA) is a key luminal actor of secretory granule biogenesis at the trans-Golgi network (TGN) level but the molecular mechanisms involved remain obscure. Here, we investigated the possibility that CgA acts synergistically with specific membrane lipids to trigger secretory granule formation. We show that CgA preferentially interacts with the anionic glycerophospholipid phosphatidic acid (PA). In accordance, bioinformatic analysis predicted a PA-binding domain (PABD) in CgA sequence that effectively bound PA (36:1) or PA (40:6) in membrane models. We identified PA (36:1) and PA (40:6) as predominant species in Golgi and granule membranes of secretory cells, and we found that CgA interaction with these PA species promotes artificial membrane deformation and remodeling. Furthermore, we demonstrated that disruption of either CgA PABD or phospholipase D (PLD) activity significantly alters secretory granule formation in secretory cells. Our findings show for the first time the ability of CgA to interact with PLD-generated PA, which allows membrane remodeling and curvature, key processes necessary to initiate secretory granule budding.


Subject(s)
Chromogranin A/metabolism , Golgi Apparatus/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/physiology , Secretory Vesicles/physiology , Animals , COS Cells , Chlorocebus aethiops , Mice , Mice, Knockout
14.
J Immunol ; 203(1): 48-57, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31109955

ABSTRACT

CD4 T cells play a critical role in promoting the development of autoimmunity in type 1 diabetes. The diabetogenic CD4 T cell clone BDC-2.5, originally isolated from a NOD mouse, has been widely used to study the contribution of autoreactive CD4 T cells and relevant Ags to autoimmune diabetes. Recent work from our laboratory has shown that the Ag for BDC-2.5 T cells is a hybrid insulin peptide (2.5HIP) consisting of an insulin C-peptide fragment fused to a peptide from chromogranin A (ChgA) and that endogenous 2.5HIP-reactive T cells are major contributors to autoimmune pathology in NOD mice. The objective of this study was to determine if poly(lactide-co-glycolide) (PLG) nanoparticles (NPs) loaded with the 2.5HIP Ag (2.5HIP-coupled PLG NPs) can tolerize BDC-2.5 T cells. Infusion of 2.5HIP-coupled PLG NPs was found to prevent diabetes in an adoptive transfer model by impairing the ability of BDC-2.5 T cells to produce proinflammatory cytokines through induction of anergy, leading to an increase in the ratio of Foxp3+ regulatory T cells to IFN-γ+ effector T cells. To our knowledge, this work is the first to use a hybrid insulin peptide, or any neoepitope, to re-educate diabetogenic T cells and may have significant implications for the development of an Ag-specific therapy for type 1 diabetes patients.


Subject(s)
Chromogranin A/metabolism , Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Insulin/metabolism , Nanoparticles/therapeutic use , Peptides/metabolism , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Genetically Modified , Cells, Cultured , Chromogranin A/genetics , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Insulin/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Nanoparticles/metabolism , Peptides/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics
15.
Bioorg Chem ; 115: 105266, 2021 10.
Article in English | MEDLINE | ID: mdl-34449322

ABSTRACT

Human Chromogranin A N46 (CGA-N46) is a weak cationic α-helical peptide with wide-spectrum antibacterial, fungal, and anticancer activities. In this study, the recombinant human CGA-N46 peptide was expressed successfully in Escherichia coli. The gene of CGA-N46 was cloned into the expression vector pET-15b without a fusion tag at the N terminus and the peptide was expressed using Isopropyl-ß-d-thiogalactoside (IPTG) as an inducer. Using 8 M guanidinium HCl, inclusion bodies containing the peptide were purified and solubilized. The rhCGA-N46 peptide was purified using Q-FF anion exchange column. The cytotoxicity of the purified rhCGA-N46 peptide was investigated on WI-38 human lung normal cell line. The anticancer activity was studied on human colon cancer cell line; HCT-116 cell line. Besides, the possible involvement of rhCGA-N46 peptide in regulating apoptotic signal pathways was analyzed by detecting the expression levels of BCL2, BID, and CAS-8 in the treated cells. The results concluded that the active peptide recovery was up to 41.98%. The purified rhCGA-N46 was safe on normal cells with IC50 = 227.74 µg/ml (40.67 µM) and had an obvious anticancer effect on colon cancer cells with IC50 = 1.997 µg/ml (356.6 nM). The expression level of BCL2 was down-regulated and BID and CAS-8 were up-regulated significantly in treated HCT-116 cells compared to untreated. In conclusion, the rhCGA-N46 peptide was produced successfully in the native form with promising anti-colon cancer activity.


Subject(s)
Chromogranin A/metabolism , Colonic Neoplasms/drug therapy , Peptides/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromogranin A/chemistry , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Peptides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship
16.
Pathol Int ; 71(10): 682-691, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34320691

ABSTRACT

Prostatic and colon carcinomas with neuroendocrine differentiation are reported to behave more aggressively than those without such differentiation. In hepatocellular carcinomas (HCCs), however, only a few studies have reported the expression status of neuroendocrine markers and somatostatin receptor 2, the main target of a somatostatin analog. Furthermore, the prognostic significance of the markers in HCCs has not been fully explored. We evaluated the expression of the neuroendocrine makers (chromogranin A, synaptophysin, and CD56) and somatostatin receptor 2 (SSTR2) in 95 HCCs, and investigated the correlation between the expression of these markers and clinicopathological findings. Chromogranin A was immunolocalized in 2 cases, synaptophysin in 15 cases, CD56 in 11 cases, and SSTR2 in 19 cases. Immunoreactivity of synaptophysin and CD56 were the significant unfavorable prognostic factors in terms of 2-year disease-free survival (DFS) and the overall survival (OS) along with a high nuclear mitosis level (>10/10 high-power field), a larger tumor size (>5 cm), the presence of vascular and/or biliary invasion, and high TNM stage (III/IV). Multivariate Cox proportional hazards analysis identified synaptophysin as an independent prognostic factor for 2-year DFS and OS. Synaptophysin expression can be used to predict an unfavorable prognosis in patients with HCC.


Subject(s)
Biomarkers, Tumor/metabolism , CD56 Antigen/metabolism , Carcinoma, Hepatocellular/diagnosis , Chromogranin A/metabolism , Liver Neoplasms/diagnosis , Receptors, Somatostatin/metabolism , Synaptophysin/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Analysis
17.
BMC Pregnancy Childbirth ; 21(1): 680, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34620125

ABSTRACT

BACKGROUND: Although preeclampsia has long been recognized as a condition affecting late pregnancy, little is known of its pathogenesis or treatment. The placenta releases a number of hormones and molecules that influence the course of pregnancy, one of which is chromogranin A, a soluble protein secreted mainly from the chromaffin cells of the adrenal medulla. Its role in pregnancy and pregnancy-related disorders remains unclear. Therefore, the main aim of the proposed study is to determine whether chromogranin A is related with the occurrence of preeclampsia. METHODS: Placental samples were collected from 102 preeclamptic patients and 103 healthy controls, and Chromogranin A gene (CHGA) expression was measured using real-time RT-PCR, The RT-PCR results were verified on the protein level using ELISA. The normal distribution of the data was tested using the Shapiro-Wilk test. The clinical and personal characteristics of the groups were compared using the Student's t-test for normally-distributed data, and the χ2 test for categorical variables. The Mann-Whitney U test was used for non-normally distributed data. As the log- transformation was not suitable for the given outcomes, the Box- Cox Transformation was used to normalize data from ELISA tests and CHGA expression. Values of P < .05 were considered statistically significant. RESULTS: Chromogranin A gene expression was found to be significantly higher in the study group than in controls. Protein analyses showed that although the CgA concentration in placental samples did not differ significantly, the catestatin (CST) level was significantly lower in samples obtained from women with preeclampsia, according to the controls. CONCLUSIONS FOR PRACTICE: This study for the first time reveals that chromogranin A gene expression level is associated with preeclampsia. Moreover, the depletion in catestatin level, which plays a protective role in hypertension development, might be a marker of developing preeclampsia. Further studies may unravel role of Chromogranin A in the discussed disease.


Subject(s)
Chromogranin A/metabolism , Peptide Fragments/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Biomarkers/metabolism , Case-Control Studies , Chromogranin A/genetics , Female , Gene Expression , Humans , Peptide Fragments/genetics , Pre-Eclampsia/genetics , Pregnancy
18.
Biol Reprod ; 102(3): 730-739, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31786596

ABSTRACT

Over the last decades, fertility of dairy cows has declined due to selection strategies focusing on milk yield. To study the effect of genetic merit for fertility on the proteome of the bovine uterine luminal fluid, Holstein heifers with low- and two groups of heifers with high-fertility index (high-fertility Holstein and Montbéliarde) were investigated. To focus on the maternal effect, heifers from all groups were synchronized and received on Day 7 high-quality embryos. Uterine luminal fluid from Day 19 pregnant heifers was analyzed in a holistic proteomic approach using nano-LC-MS/MS analysis combined with a label-free quantification approach. In total, 1737 proteins were identified, of which 597 differed significantly in abundance between the three groups. The vast majority of proteome differences was found comparing both high-fertility groups to the low-fertility Holstein group, showing that the genetic predisposition for fertility is prevalent regarding the uterine luminal fluid proteome. Evaluation of this dataset using bioinformatic tools revealed an assignment of higher abundant proteins in low-fertility Holstein to several metabolic processes, such as vitamin metabolic process, which comprises folate receptor alpha (FOLR1) and retinol-binding protein, indicating an involvement of disturbed metabolic processes in decreased fertility. Moreover, immune system-related proteins - lactotransferrin and chromogranin A - were enriched in low-fertility cows together with interferon tau 3 h and interferon tau-2. Our results indicate that the genetic merit for fertility leads to substantial quantitative differences at the level of proteins in uterine fluid of pregnant animals, thus altering the microenvironment for the early conceptus.


Subject(s)
Fertility/physiology , Proteome/metabolism , Uterus/metabolism , Animals , Cattle , Chromogranin A/metabolism , Computational Biology , Female , Folate Receptor 1/metabolism , Lactoferrin/metabolism , Proteomics , Tandem Mass Spectrometry
19.
Cell Tissue Res ; 379(1): 157-167, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31673758

ABSTRACT

REIC (reduced expression in immortalized cells) has been identified as a gene whose expression was reduced in immortalized cultured cells. The REIC gene is identical to Dickkopf-3 (Dkk3), which encodes a secreted glycoprotein belonging to the Dkk family. Previously, we showed that Dkk3 protein is present in the mouse adrenal medulla. However, its role in this tissue has not been elucidated. To explore it, we performed electron microscopic (EM) studies and RNA-sequencing (RNA-seq) analysis on Dkk3-null adrenal glands. EM studies showed that the number of dense core secretory vesicles were significantly reduced and empty vesicles were increased in the medulla endocrine cells. Quantitative PCR (qPCR) analysis showed relative expression levels of chromogranin A (Chga) and neuropeptide Y (Npy) were slightly but significantly reduced in the Dkk3-null adrenal glands. From the result of RNA-seq analysis as a parallel study, we selected three of the downregulated genes, uncoupled protein-1 (Ucp1), growth arrest and DNA-damage-inducible 45 gamma (Gadd45g), and Junb with regard to the estimated expression levels. In situ hybridization confirmed that these genes were regionally expressed in the adrenal gland. However, expression levels of these three genes were not consistent as revealed by qPCR. Thus, Dkk3 maintains the integrity of secreting vesicles in mouse adrenal medulla by regulating the expression of Chga and Npy.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Adrenal Medulla/physiology , Secretory Vesicles/physiology , Adaptor Proteins, Signal Transducing/genetics , Adrenal Medulla/cytology , Adrenal Medulla/ultrastructure , Animals , Chromogranin A/metabolism , Down-Regulation , Female , In Situ Hybridization , Mice , Mice, Knockout , Neuropeptide Y/metabolism , RNA, Messenger , RNA-Seq , Secretory Vesicles/ultrastructure , Transcriptome
20.
BMC Cancer ; 20(1): 27, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31924180

ABSTRACT

BACKGROUND: High grade gastroenteropancreatic (GEP) neuroendocrine neoplasms (NEN) with a Ki67 proliferation index > 20%, include well-differentiated tumours grade 3 (NET G3) and poorly differentiated (PD) neuroendocrine carcinomas (NEC). Abnormal p53-expression is a feature of PD tumours, while expression of chromogranin A (CgA) and somatostatin-receptor 2a (SSTR-2a) may be a feature of well-differentiated tumours. The aim of this study was to elucidate the expression and prognostic value of these three markers in 163 GEP-NEN patients with a Ki67-index > 20%. METHOD: Clinical data, histopathology and overall survival were analysed according to Kaplan-Meier's method and Cox regression. The expression of SSTR-2a, CgA and synaptophysin was analysed in tumour specimens by immunohistochemistry, and semi-quantitatively scored as negative (< 5%), heterogeneously positive (5-30%) or strongly positive (> 30%). P53 was defined as normal when scored as heterogeneously positive (1-30%), and abnormal when negative (0%) or strongly positive (> 30%). RESULTS: In multivariate analysis, better survival was observed among patients with heterogeneously positive p53 compared to strongly positive (p < 0.001). When dichotomised, tumours with a heterogeneously positive p53 vs. negative and strongly positive p53 also showed a significantly better survival (p = 0.002). Survival was significantly worse for negative CgA compared to heterogeneously positive CgA (p = 0.02). Strongly positive SSTR-2a expression was found in 26% of the 163 included patients. Well-differentiated morphology correlated with strong expression of SSTR-2a and CgA, and heterogeneously positive p53-staining, and was more frequent in pancreatic primaries. In pancreatic primaries, strongly positive SSTR-2a was associated with longer survival (univariate analysis, p = 0.02). A significantly lower Ki67 proliferation index was found in patients with a heterogeneously positive p53, a positive SSTR-2a and CgA expression. CONCLUSION: Our results suggest that abnormal p53-expression is an independent negative prognostic marker in GEP-NEN with a Ki67-index > 20%. Patients with heterogeneously positive p53 had the best prognosis. SSTR-2a was a positive prognostic marker in pancreatic NEN. Negative CgA was associated with a significantly worse OS compared to heterogeneously positive CgA-expression in a multivariate sub-analysis. Lower Ki67 index correlated significantly with heterogeneously positive p53, positive SSTR-2a and CgA expression.


Subject(s)
Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Chromogranin A/metabolism , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Carcinoma, Neuroendocrine/etiology , Carcinoma, Neuroendocrine/mortality , Cell Line, Tumor , Female , Follow-Up Studies , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/mortality , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Grading , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/mortality , Prognosis , Proportional Hazards Models , Tumor Suppressor Protein p53/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL