Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cardiovasc Pathol ; 70: 107630, 2024.
Article in English | MEDLINE | ID: mdl-38490313

ABSTRACT

A female neonate born with normal Apgar scores at 38+2 weeks of gestational age unexpectedly passed away within less than 30 hours after birth. The situation mirrored her brother's earlier demise within 24 hours post-delivery, suggesting a possible genetic disorder. Gross examination revealed widespread cyanosis and distinct yellowish changes on the cardiac ventricles. Histopathological examination disclosed lipid accumulation in the liver, heart, and kidneys. Tandem mass spectrometry detected elevated levels of 10 amino acids and 14 carnitines in cardiac blood. Trio-whole genome sequencing (Trio-WGS) identified the SLC25A20 c.199-10T>G mutation associated with carnitine-acylcarnitine translocase disease (CACTD), a type of fatty acid oxidation disorders (FAODs) with a potential for sudden death. Further validation of gene expression confirmed the functional deficiency of SLC25A20, ultimately diagnosing CACTD as the underlying cause of the neonate's demise. This case highlights the importance of prenatal metabolic and genetic screening for prospective parents and emphasizes the need for forensic doctors to integrate metabolomic and genomic investigations into autopsies for suspected inherited metabolic diseases.


Subject(s)
Carnitine Acyltransferases , Lipid Metabolism, Inborn Errors , Mutation , Humans , Infant, Newborn , Female , Carnitine Acyltransferases/deficiency , Carnitine Acyltransferases/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/diagnosis , Phenotype , Fatal Outcome , Genetic Predisposition to Disease , Sudden Infant Death/genetics , Sudden Infant Death/pathology , Sudden Infant Death/etiology , Autopsy , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Cause of Death , Carnitine/analogs & derivatives , Carnitine/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/pathology , Myocardium/metabolism , Membrane Transport Proteins
2.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1013477

ABSTRACT

@#目的:探讨LINC00958/血管内皮生长因子C(VEGF-C)信号通路在宫颈癌的淋巴管生成和淋巴转移中的作用。方法:从2020年9月至2022年9月期间在河南省人民医院接受手术的患者中收集了42例宫颈癌组织标本,通过qPCR检测宫颈癌组织和宫颈癌细胞(Hela、C33A、SiHa、Caski)中LINC00958的表达情况。将LINC00958过表达载体(LINC00958组)或对照载体(CMV组)转染Caski细胞,敲减LINC00958(shLINC00958组)、VEGF-C(shVEGF-C组)的shRNA序列或阴性对照shRNA(shNC组)转染SiHa细胞。分别通过CCK-8法、Transwell实验检测过表达或敲减LINC00958对宫颈癌细胞增殖、迁移和侵袭的影响。观察转染后细胞的培养上清液对人淋巴管内皮细胞(HLEC)淋巴管形成能力的影响。建立小鼠腘淋巴结转移模型,观察过表达LINC00958或同时敲减VEGF-C对宫颈癌淋巴结转移的影响。结果:LINC00958在宫颈癌组织中呈高表达(P<0.001),高水平的LINC00958与大肿瘤、晚期肿瘤分级、浸润深度和淋巴转移有关联(P<0.05或P<0.01)。与正常人宫颈上皮细胞ende1617相比,宫颈癌细胞中LINC00958水平均显著升高(P<0.01或P<0.001)。shLINC00958组SiHa细胞的增殖、迁移、侵袭能力及其培养上清液的促HLEC淋巴管形成能力均显著低于shNC组(P<0.05、P<0.01或P<0.001),LINC00958组Caski细胞的增殖、迁移、侵袭能力及其培养上清液的促HLEC淋巴管形成能力显著高于CMV组(P<0.05、P<0.01或P<0.001)。通过RNA下拉、RNA免疫沉淀实验发现宫颈癌细胞中LINC00958能够特异性结合VEGF-C。LINC00958+shVEGF-C组Caski细胞的增殖、迁移、侵袭能力及其培养上清液的促淋巴管形成能力显著低于LINC00958组(P<0.01或P<0.001);在小鼠腘淋巴结转移模型中,LINC00958+shVEGF-C组中小鼠腘窝淋巴结的体积和VEGF-C蛋白、N-cadherin蛋白以及LYVE-1的阳性细胞比例均显著低于LINC00958组(均P<0.001)。结论:LINC00958通过直接与VEGF-C蛋白相互作用增强宫颈癌细胞的增殖、侵袭、淋巴管生成能力,促进小鼠腘淋巴结转移模型的淋巴结转移。

3.
Forensic Sci Int ; 354: 111912, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38103455

ABSTRACT

INTRODUCTION: Objective assessment of cardiac hypertrophy in forensic pathology practice is of great significance for forensic pathologists, for whom reference values for normal heart weights are needed. Developed regions such as Europe, the United States, and Japan recalculate the weight of human organs at regular intervals, but in China, there has been no systematic calculation of the weights of human organs since 2006. AIMS: To statistically analyse the heart weight of Chinese adults postmortem and obtain a reference range. MATERIALS AND METHODS: 4170 adult autopsy reports were collected from 12 forensic departments in 10 provinces in China. The causes of death were classified by sex, and heart weight and the heart weight/body height ratio reference values were further calculated according to different body mass index and body heights. Finally, the cutoff value of cardiac hypertrophy in Chinese adults was calculated. RESULTS: In the group of non-cardiovascular disease causes of death, the cardiac weight of the electric death group was higher, while the heart weight of the prolonged bed-rest group was significantly reduced. After the electric death and prolonged bed-rest groups were excluded, heart weight, the heart weight/body height ratio, and cutoff values for cardiac hypertrophy were further classified and analysed according to body mass index. The mean reference values for heart weight in men and women with normal weight status were 325.82 ± 41.60 g and 286.39 ± 44.84 g, and the heart weight/body height ratios were 1.95 ± 0.23 in men and 1.82 ± 0.27, respectively. The cutoff values for cardiac hypertrophy were 387.35 g for men and 346.80 g for women. CONCLUSION: The heart weight reference values of both sexes in this study were significantly higher than those in 2006, which is considered related to the development of China's economy and the improvement of people's living standards. This study also suggests the need for a new round of statistical surveys and updated data on the weight of other organs.


Subject(s)
Cardiomegaly , Heart , Male , Adult , Humans , Female , Autopsy , Forensic Pathology , China , Body Weight , Organ Size
4.
J Cancer ; 14(9): 1648-1659, 2023.
Article in English | MEDLINE | ID: mdl-37325054

ABSTRACT

Cervical cancer (CC) is one of the leading cancers among the female reproductive system. The piwi-interacting RNA (piRNA) function and biogenesis has been studied in various cancers, including CC. But the precise mechanism of piRNA in CC is still unknown. In our study, we found that piRNA-17458 was overexpressed in CC tissues and cells. piRNA-17458 mimic and inhibitor promoted and suppressed proliferation, migration and invasion ability of CC cells, respectively. We also demonstrated that piRNA-17458 mimic could contribute to tumor growth in mice xenograft models. Besides, we also found that the piRNA-17458 mimic could enhance mRNA N6-methyladenosine(m6A) levels and increase WTAP stability in CC cells, while the effects of the mimic was reversed by the WTAP knockdown. The results of dual luciferase reporter assay showed that WTAP was a direct target of piRNA-17458. Knockdown of WTAP attenuated proliferation, migration and invasion of CC cells in piRNA-17458 mimic group. Our finding not only demonstrates for the first time that piRNA-17458 is overexpressed in CC tissues and cells, but also shows that piRNA-17458 promotes tumorigenesis of CC in a WTAP-mediated m6A methylation manner.

6.
Reprod Biol ; 22(1): 100592, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34995817

ABSTRACT

Endometriosis is a benign gynecological disease. Accumulating evidence has revealed the participation of dysregulated miRNAs in the progression of endometriosis. Here, the function and molecular mechanism of miR-143-3p in endometriosis were investigated. The levels of vasohibin 1 (VASH1) and miR-143-3p in endometrial tissues and endometriotic stromal cells (ESCs) were detected by RT-qPCR. Migrative and invasive phenotypes of ESCs were tested by Transwell assays. The protein expression of VASH1, TGF-ß signaling markers, and epithelial to mesenchymal transition (EMT) markers was examined by western blotting. The targeted relationship between miR-143-3p and VASH1 was confirmed by bioinformatics analysis and luciferase reporter assay. We found that miR-143-3p expression was significantly upregulated in ectopic endometrial tissues compared to that in eutopic and normal endometrial tissues. MiR-143-3p knockdown restrained EMT process, invasive and migrative behaviors of ESCs. Mechanically, miR-143-3p targeted VASH1 and negatively regulated VASH1. VASH1 downregulation reserved the effects of miR-143-3p knockdown in ESCs. MiR-143-3p activated TGF-ß signaling via targeting VASH1. Furthermore, activation of TGF-ß signaling counteracted the miR-143-3p knockdown-caused suppression of migration, invasion and EMT process in ESCs. Overall, miR-143-3p activates TGF-ß signaling by targeting VASH1 to facilitate migration and invasion of ESCs.


Subject(s)
Endometriosis , MicroRNAs , Stromal Cells , Cell Cycle Proteins/metabolism , Cell Movement/genetics , Endometriosis/genetics , Endometriosis/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism
7.
Transl Cancer Res ; 10(7): 3306-3316, 2021 Jul.
Article in English | MEDLINE | ID: mdl-35116637

ABSTRACT

BACKGROUND: Pancreatic cancer is generally characterized with high levels of malignancy and poor prognosis. In addition, there are currently no effective therapeutic agents against the disease. However, apatinib which is a small molecular agent targeting the vascular endothelial growth factor receptor 2 (VEGFR-2), has been shown to generate favorable outcomes in gastric cancer. Therefore, the present study explored the effects of apatinib on pancreatic cancer. METHODS: The activity of the ASPC-1 or PANC-1 cells was examined through colony formation assays, wound healing experiments as well as the Transwell and Western blot (WB) analyses. Additionally, a xenograft model was established by subcutaneously injecting the ASPC-1 cells into nude mice. Microvessel density (MVD) and Ki-67 expression were examined through immunohistochemistry (IHC) and WB analyses. RESULTS: The findings showed that treatment with either 10 or 20 µM of apatinib led to a decrease in the proliferation, migration and invasion of ASPC-1 and PANC-1 cells. Additionally, apatinib significantly hindered xenograft growth. Moreover, there was a decrease in Ki-67 expression and MVD, 21 days after treatment with apatinib. The results also showed that apatinib had no effect on the levels of the VEGFR-2, ERK1/2 and AKT proteins although there was a significant decrease in the expression of phosphate VEGFR2 (p-VEGFR2), phosphate AKT (p-AKT) and phosphate ERK1/2 (p-ERK1/2). CONCLUSIONS: Apatinib inhibits the proliferation and migration of pancreatic cancer cells, blocking growth and angiogenesis in transplanted tumors. In addition, the underlying mechanism may involve phosphorylation of the PI3K/AKT and ERK1/2/MAPKs signaling pathways.

8.
Ther Adv Med Oncol ; 12: 1758835920940932, 2020.
Article in English | MEDLINE | ID: mdl-32728393

ABSTRACT

BACKGROUND: The evidence base for optimum third-line therapy for metastatic colorectal cancer (mCRC) is not conclusive. Recent studies have demonstrated the efficacy of regorafenib as third-line therapy in mCRC. This indirect meta-analysis compared the efficacy and safety of regorafenib with other available third-line therapies for mCRC. METHODS: A literature search for randomized controlled trials (RCTs) was conducted in PubMed, Embase, and Cochrane Library for studies evaluating the efficacy and safety of fruquintinib, regorafenib, TAS-102, and nintedanib as third-line therapies in patients with mCRC. Overall survival (OS) and progression-free survival (PFS) were the primary outcomes, while objective response rate (ORR) and safety were the secondary outcomes. Hazard ratio (HR) and relative risk (RR) with their respective 95% confidence interval (CI) were used for analysis of survival, clinical response, and safety data. An adjusted indirect meta-analysis with placebo as the common comparator was performed. RESULTS: We identified eight RCTs comparing regorafenib (two studies), fruquintinib (two studies), TAS-102 (three studies), and nintedanib (one study) against placebo. The OS with regorafenib was significantly better when compared with nintedanib (HR = 0.66; 95% CI: 0.45, 0.95, p = 0.02) but was similar to that of fruquintinib (HR = 1.01; 95% CI: 0.67, 1.52, p = 0.94) and TAS-102 (HR = 0.97; 95% CI: 0.68, 1.38, p = 0.88). The PFS and ORR for regorafenib were slightly better than those of TAS-102 (PFS: HR = 0.86, 95% CI: 0.54, 1.37, p = 0.5; ORR: RR = 1.13, 95% CI: 0.11, 11.05, p = 0.92) and nintedanib (PFS: HR = 0.68, 95% CI: 0.42, 1.10, p = 0.12; ORR: not reported) but were lower than those for fruquintinib (PFS: HR = 1.53, 95% CI: 0.93, 2.52, p = 0.08; ORR: RR = 0.68269, 95% CI: 0.045, 10.32, p = 0.79). Safety analysis showed that the RR of adverse events (AEs) was lesser in patients treated with regorafenib in comparison with that in patients treated with fruquintinib, but was similar to that in patients treated with nintedanib and TAS-102. CONCLUSION: Regorafenib has efficacy similar to that of TAS-102 and better safety when compared with fruquintinib. Considering the mechanism of action of regorafenib, which targets multiple factors in the angiogenic pathway, it could be an ideal option for treatment in the beyond second-line setting.

9.
J Cancer ; 11(1): 25-40, 2020.
Article in English | MEDLINE | ID: mdl-31892970

ABSTRACT

Chemotherapy resistance represents a major obstacle for the treatment of patients with breast cancer (BC) and greatly restricts the therapeutic effect of the first-line chemotherapeutic agent doxorubicin (DOX). The present study aimed to investigate the feasibility of the recombinant dual-target murine double minute 2 (MDM2) and murine double minute X (MDMX) inhibitor in reversing the DOX resistance of BC. Both DOX-resistant human breast carcinoma cell lines exhibited a multidrug resistance (MDR) phenotype. The ability of the dual-target MDM2/MDMX inhibitor in reversing doxorubicin resistance was subsequently verified, (9.15 and 13.92 - fold reversal indexes) respectively. We observed that the MDM2/MDMX inhibitor in combination with DOX could suppress proliferation, promote cell cycle arrest and induce apoptosis. In addition, it was capable of reducing rhodamine123 efflux in DOX-resistance BC cell lines and further played a key role in BC nude mice model. The groups that were treated with the combination of the drugs had decreased P-glycoprotein/multidrug resistance-associated protein/cdc 2/Bcl-2 expression and increased CyclinB1/Bax expression. These effects were caused due to activation of the transforming growth factor ß-activated kinase 1 (TAK1)-binding protein 1 (TAB1)/TAK1/p38 mitogen-activated protein kinase (MAPK) signaling pathway, as shown by small interfering RNA (siRNA) silencing and immumohistochemical staining of BC tissue sections. Furthermore, high MDM2/MDMX expression was positively associated with weak TAB1 expression in BC patients. Therefore, the recombinant dual-target MDM2/MDMX inhibitor could reverse doxorubicin resistance via the activation of the TAB1/TAK1/p38 MAPK pathway in wild-type p53 multidrug-resistant BC.

10.
J Cell Mol Med ; 24(1): 760-771, 2020 01.
Article in English | MEDLINE | ID: mdl-31762191

ABSTRACT

High rates of de novo lipid synthesis have been discovered in certain kinds of tumours, including gallbladder cancer (GBC). Unlike several other tumours, GBC is highly insensitive to standard adjuvant therapy, which makes its treatment even more challenging. Although several potential targets and signalling pathways underlying GBC chemoresistance have been revealed, the precise mechanisms are still elusive. In this study, we found that α-Mangostin, as a dietary xanthone, repressed the proliferation and clone formation ability, induced cell cycle arrest and the apoptosis, and suppressed de novo lipogenesis of gallbladder cancer cells. The underlying mechanisms might involve the activation of AMPK and, therefore, the suppression of SREBP1 nuclear translocation to blunt de novo lipogenesis. Furthermore, SREBP1 silencing by siRNA or α-mangostin enhanced the sensitivity of gemcitabine in gallbladder cancer cells. In vivo studies also displayed that MA or gemcitabine administration to nude mice harbouring NOZ tumours can reduce tumour growth, and moreover, MA administration can significantly potentiate gemcitabine-induced inhibition of tumour growth. Corroborating in vitro findings, tumours from mice treated with MA or gemcitabine alone showed decreased levels of proliferation with reduced Ki-67 expression and elevated apoptosis confirmed by TUNEL staining, furthermore, the proliferation inhibition and apoptosis up-regulation were obviously observed in MA combined with gemcitabine treatment group. Therefore, inhibiting de novo lipogenesis via targeting the AMPK/SREBP1 signalling by MA might provide insights into a potential strategy for sensitizing GBC cells to chemotherapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Deoxycytidine/analogs & derivatives , Drug Synergism , Gallbladder Neoplasms/drug therapy , Lipogenesis/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Xanthones/pharmacology , AMP-Activated Protein Kinases/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints , Cell Movement , Cell Proliferation , Deoxycytidine/pharmacology , Drug Therapy, Combination , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Sterol Regulatory Element Binding Protein 1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
11.
Biochem Pharmacol ; 169: 113610, 2019 11.
Article in English | MEDLINE | ID: mdl-31465777

ABSTRACT

The incidence of melanoma is increasing rapidly worldwide. Additionally, new and effective candidates for treating melanoma are needed because of the increase in drug resistance and the high metastatic potential of this cancer. The STAT3 signaling pathway plays a pivotal role in pathogenesis of melanoma, making STAT3 a promising anticancer target for melanoma therapy. Niclosamide, an FDA-approved anti-helminthic drug, has been identified as a potent STAT3 inhibitor that suppresses STAT3 phosphorylation at Tyr705 and its transcript activity. In this study, we evaluated the biological activities of niclosamide in melanoma in vitro and in vivo. Niclosamide potently inhibited the growth of four melanoma cell lines and induced the apoptosis of melanoma cells via the mitochondrial apoptotic pathway. Further, western blot analysis indicated that cell apoptosis was correlated with activation of Bax and cleaved caspase-3 and decreased expression of Bcl-2. Moreover, niclosamide markedly impaired melanoma cell migration and invasion, reduced phosphorylated STAT3Tyr705 levels, and inhibited matrix metalloproteinase-2 and -9 expression. Additionally, in a xenograft model of A375, intraperitoneal administration of niclosamide inhibited tumor growth and tumor weight in a dose-dependent manner without obvious side effects. Histological and immunohistochemical analyses revealed a decrease in Ki-67-positive cells and p-STAT3Try705-positive cells and increase in cleaved caspase-3-positive cells. Notably, niclosamide significantly inhibited pulmonary metastasis in a B16-F10 melanoma lung metastasis model, including the number of lung metastatic nodules and lung/body coefficient. Importantly, a marked reduction in myeloid-derived suppressor cells (Gr1+CD11b+) infiltration in the pulmonary metastasis tissue was observed. Taken together, these results demonstrate that niclosamide is a promising candidate for treating melanoma.


Subject(s)
Anthelmintics/pharmacology , Drug Repositioning , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Niclosamide/pharmacology , STAT3 Transcription Factor/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Lung Neoplasms/prevention & control , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/physiology , Niclosamide/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Cancer Biol Ther ; 20(5): 617-632, 2019.
Article in English | MEDLINE | ID: mdl-30462562

ABSTRACT

Triple-negative breast cancer (TNBC) has a poor prognosis mainly due to insensitivity or resistance to standard anthracycline- and taxane-based chemotherapy, urgently calling for new adjuvants to reverse drug resistance. Dual-target murine double minute 2 (MDM2) and murine double minute X (MDMX) inhibitor has been proved to play a critical part against cancer, particularly focusing on the tremendous potential to enhance the efficacy of doxorubicin (DOX), however little was reported in TNBC. In the present study, we investigated the synergistic antitumor effect of the MDM2/MDMX inhibitor with DOX using three TNBC cell lines, two in situ transplantation tumor models and 214 clinical samples. We observed that the MDM2/MDMX inhibitor combined with DOX could not only inhibit cell vitality and migration and invasion abilities, but also highly inhibit tumor growth in TNBC nude mice. Besides, co-treatment of MDM2/MDMX inhibitor and DOX suppressed epithelial to mesenchymal transition (EMT) through increasing the TAK1-binding protein 1 (TAB1), transforming growth factor ß-activated kinase 1 (TAK1) and p38 mitogen-activated protein kinase (MAPK) expression. Small interfering RNA-mediated TAB1 knockdown induced the EMT, desensitized cells to DOX and enhanced the migration and invasion abilities. High MDM2/MDMX expression was positively associated with weak TAB1 expression in 214 TNBC tumor tissues confirmed by immumohistochemical staining and MDM2/MDMX/TAB1 expression was significantly related to TNBC patient survival. These findings indicate that dual-target MDM2/MDMX inhibitor could increase the sensitization of doxorubicin and inhibit migration and invasion abilities in TNBC cells through p38 MAPK pathway activation caused EMT suppression and hence could be useful in TNBC treatments in future.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Doxorubicin/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aftercare , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/pathology , Breast/surgery , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Retrospective Studies , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/surgery , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...