Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 840210, 2022.
Article in English | MEDLINE | ID: mdl-35531327

ABSTRACT

In the summer of 2019, DiaSorin Molecular started designing a multiplex respiratory panel with pan-coronavirus detection as one of the planned targets. The R&D team in Gerenzano, Italy was already searching databases, performing alignments and assessing preliminary target regions for common coronavirus RT-PCR, including SARS and MERS-CoV. In December 2019, we were vigilant and following a cluster of pneumonia cases with undetermined etiology in Wuhan, China. As we now know, the cause of the respiratory infections was the new SARS-CoV-2 virus. DiaSorin Molecular swiftly responded in line with our heritage and company history in detecting emerging infectious diseases. Early in the pandemic and in record time, using research and development teams in both Italy and the U.S. together with the U.S. manufacturing team, we were able to develop and commercialize a new diagnostic test, Simplexa™ COVID-19 Direct, to detect SARS-CoV-2. Our unique platform allowed development of a rapid diagnostic test without the need for extraction reagents. Challenges with control materials, quarantines, clinical samples, raw materials and production were overcome and the entire company worked side by side for accelerated delivery of this assay to clinical labs in Europe, the U.S. and Canada.


Subject(s)
COVID-19 , COVID-19/diagnosis , COVID-19 Testing , China , Humans , Pandemics , SARS-CoV-2/genetics , Sensitivity and Specificity
2.
Expert Rev Mol Diagn ; 16(3): 269-76, 2016.
Article in English | MEDLINE | ID: mdl-26679835

ABSTRACT

The Simplexa™ Group A Strep Direct assay is intended for use on the Integrated Cycler for detection of Group A Streptococcus (GAS) directly from throat swabs that have not undergone nucleic acid extraction. A prospective study of 1352 samples in 4 geographically diverse sites showed an overall prevalence of GAS of 15.4%. The assay demonstrated 97.4% sensitivity and 95.2% specificity versus culture. The positive predictive value compared to culture was 72.7%. However, 46 out of 57 discrepant samples were Group A Strep positive when tested using a bi-directional sequencing method illustrating the increased sensitivity of the assay compared to culture for detection of GAS. Rapid and accurate diagnosis of GAS allows for timely treatment to decrease complications of this prevalent organism that continues to cause substantial morbidity and mortality worldwide.


Subject(s)
Molecular Diagnostic Techniques/methods , Pharyngitis/diagnosis , Streptococcal Infections/diagnosis , Streptococcus pyogenes/isolation & purification , Humans , Molecular Diagnostic Techniques/instrumentation , Pharyngitis/microbiology , Polymerase Chain Reaction/instrumentation , Polymerase Chain Reaction/methods , Sensitivity and Specificity , Streptococcus pyogenes/genetics
3.
BMC Cancer ; 9: 3, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-19123943

ABSTRACT

BACKGROUND: The steroid and xenobiotic receptor, SXR, is an orphan nuclear receptor that regulates metabolism of diverse dietary, endobiotic, and xenobiotic compounds. SXR is expressed at high levels in the liver and intestine, and at lower levels in breast and other tissues where its function was unknown. Since many breast cancer preventive and therapeutic compounds are SXR activators, we hypothesized that some beneficial effects of these compounds are mediated through SXR. METHODS: To test this hypothesis, we measured proliferation of breast cancer cells in response to SXR activators and evaluated consequent changes in the expression of genes critical for proliferation and cell-cycle control using quantitative RT-PCR and western blotting. Results were confirmed using siRNA-mediated gene knockdown. Statistical analysis was by t-test or ANOVA and a P value < or = 0.05 was considered to be significant. RESULTS: Many structurally and functionally distinct SXR activators inhibited the proliferation of MCF-7 and ZR-75-1 breast cancer cells by inducing cell cycle arrest at the G1/S phase followed by apoptosis. Decreased growth in response to SXR activation was associated with stabilization of p53 and up-regulation of cell cycle regulatory and pro-apoptotic genes such as p21, PUMA and BAX. These gene expression changes were preceded by an increase in inducible nitric oxide synthase and nitric oxide in these cells. Inhibition of iNOS blocked the induction of p53. p53 knockdown inhibited up-regulation of p21 and BAX. We infer that NO is required for p53 induction and that p53 is required for up-regulation of cell cycle regulatory and apoptotic genes in this system. SXR activator-induced increases in iNOS levels were inhibited by siRNA-mediated knockdown of SXR, indicating that SXR activation is necessary for subsequent regulation of iNOS expression. CONCLUSION: We conclude that activation of SXR is anti-proliferative in p53 wild type breast cancer cells and that this effect is mechanistically dependent upon the local production of NO and NO-dependent up-regulation of p53. These findings reveal a novel biological function for SXR and suggest that a subset of SXR activators may function as effective therapeutic and chemo-preventative agents for certain types of breast cancers.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Steroid/metabolism , Breast Neoplasms/genetics , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pregnane X Receptor , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Steroid/agonists , Receptors, Steroid/biosynthesis , Receptors, Steroid/genetics , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
4.
Environ Health Perspect ; 116(7): 880-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18629309

ABSTRACT

BACKGROUND: Nuclear receptor subfamily 1, group I, member 2 (NR1I2), commonly known as steroid and xenobiotic receptor (SXR) in humans, is a key ligand-dependent transcription factor responsible for the regulation of xenobiotic, steroid, and bile acid metabolism. The ligand-binding domain is principally responsible for species-specific activation of NR1I2 in response to xenobiotic exposure. OBJECTIVES: Our objective in this study was to create a common framework for screening NR1I2 orthologs from a variety of model species against environmentally relevant xenobiotics and to evaluate the results in light of using these species as predictors of xenobiotic disposition and for assessment of environmental health risk. METHODS: Sixteen chimeric fusion plasmid vectors expressing the Gal4 DNA-binding domain and species-specific NR1I2 ligand-binding domain were screened for activation against a spectrum of 27 xenobiotic compounds using a standardized cotransfection receptor activation assay. RESULTS: NR1I2 orthologs were activated by various ligands in a dose-dependent manner. Closely related species show broadly similar patterns of activation; however, considerable variation to individual compounds exists, even among species varying in only a few amino acid residues. CONCLUSIONS: Interspecies variation in NR1I2 activation by various ligands can be screened through the use of in vitro NR1I2 activation assays and should be taken into account when choosing appropriate animal models for assessing environmental health risk.


Subject(s)
Receptors, Steroid/biosynthesis , Xenobiotics/toxicity , Amino Acid Sequence , Animals , Cloning, Molecular , Dose-Response Relationship, Drug , Humans , Models, Animal , Molecular Sequence Data , Pregnane X Receptor , Receptors, Steroid/genetics , Species Specificity
5.
Cancer ; 109(5): 957-65, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17279585

ABSTRACT

BACKGROUND: Approximately 30% to 40% of all patients with osteosarcomas ultimately experience recurrence. The study investigated the hypothesis that the resistance of osteosarcoma to chemotherapy may be related to the expression of a pregnane xenobiotic receptor (PXR) variant protein and its role as the major inducer of P450 3A4 in these tumors. METHODS: Polymerase chain reaction (PCR) and Western blot analysis were used to determine PXR mRNA and protein expression, respectively. Real-time PCR and CYP3A catalytic activity using 7-benzyl-trifluoromethyl coumarin (BFC) as the probe substrate were used to measure the induction of P450 3A4 or MDR1. siRNA transfections were performed for PXR and cytotoxicity determined by a colorimetric based assay or Annexin v-Fitc staining. RESULTS: Differences were observed in the molecular size of the PXR protein expressed in sarcoma cell lines when compared with the wildtype PXR expressed in normal liver, kidney, or small intestine. A polyclonal PXR antibody raised against the N-terminus of the wildtype PXR did not detect PXR expressed in these sarcoma cell lines. In the osteosarcoma cell lines, etoposide and doxorubicin were better inducers of P450 3A4 and MDR1 than rifampin. siRNA against PXR down-regulated P450 3A4 expression only in the osteosarcoma cell line. Cytotoxicity assays showed that the resistance of the osteosarcoma cell lines to etoposide correlated with PXR protein expression levels and activation of P450 3A4 and could be prevented by ketoconazole. CONCLUSION: The results suggest that PXR plays a critical role in the regulation of P450 3A4 expression in osteosarcoma and that its expression and activation in these tumors may influence the effect of chemotherapeutic agents on the induction of target genes implicated in drug resistance.


Subject(s)
Drug Resistance, Neoplasm/physiology , Osteosarcoma/metabolism , Protein Isoforms/metabolism , Receptors, Steroid/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , Humans , Osteosarcoma/genetics , Pregnane X Receptor , Protein Isoforms/drug effects , RNA, Messenger/analysis , RNA, Small Interfering , Receptors, Steroid/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Rifampin/pharmacology
6.
J Clin Invest ; 116(8): 2280-2289, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16841097

ABSTRACT

While it has long been known that inflammation and infection reduce expression of hepatic cytochrome P450 (CYP) genes involved in xenobiotic metabolism and that exposure to xenobiotic chemicals can impair immune function, the molecular mechanisms underlying both of these phenomena have remained largely unknown. Here we show that activation of the nuclear steroid and xenobiotic receptor (SXR) by commonly used drugs in humans inhibits the activity of NF-kappaB, a key regulator of inflammation and the immune response. NF-kappaB target genes are upregulated and small bowel inflammation is significantly increased in mice lacking the SXR ortholog pregnane X receptor (PXR), thereby demonstrating a direct link between SXR and drug-mediated antagonism of NF-kappaB. Interestingly, NF-kappaB activation reciprocally inhibits SXR and its target genes whereas inhibition of NF-kappaB enhances SXR activity. This SXR/PXR-NF-kappaB axis provides a molecular explanation for the suppression of hepatic CYP mRNAs by inflammatory stimuli as well as the immunosuppressant effects of xenobiotics and SXR-responsive drugs. This mechanistic relationship has clinical consequences for individuals undergoing therapeutic exposure to the wide variety of drugs that are also SXR agonists.


Subject(s)
Inflammation/physiopathology , NF-kappa B/physiology , Receptors, Steroid/physiology , Steroids/metabolism , Xenobiotics/metabolism , Carcinoma, Hepatocellular , Cell Line, Tumor , Cytochrome P-450 CYP3A/metabolism , Gene Expression Regulation , Hepatocytes/physiology , Humans , Liver Neoplasms , NF-kappa B/antagonists & inhibitors , Pregnane X Receptor , Receptors, Steroid/genetics , Tumor Necrosis Factor-alpha/pharmacology
7.
Mol Endocrinol ; 20(3): 475-82, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16037129

ABSTRACT

Endocrine-disrupting chemicals (EDC) are commonly considered to be compounds that mimic or block the transcriptional activation elicited by naturally circulating steroid hormones by binding to steroid hormone receptors. For example, the Food Quality Protection Act of 1996 defines EDC as those, that "may have an effect in humans that is similar to an effect produced by a naturally occurring estrogen, or other such endocrine effect as the Administrator may designate." The definition of EDC was later expanded to include those that act on the estrogen, androgen, and thyroid hormone receptors. In this minireview, we discuss new avenues through which xenobiotic chemicals influence these and other hormone-dependent signaling pathways. EDC can increase or block the metabolism of naturally occurring steroid hormones and other xenobiotic chemicals by activating or antagonizing nuclear hormone receptors. EDC affect the transcriptional activity of nuclear receptors by modulating proteasome-mediated degradation of nuclear receptors and their coregulators. Xenobiotics and environmental contaminants can act as hormone sensitizers by inhibiting histone deacetylase activity and stimulating mitogen-activated protein kinase activity. Some endocrine disrupters can have genome-wide effects on DNA methylation status. Others can modulate lipid metabolism and adipogenesis, perhaps contributing to the current epidemic of obesity. Additional elucidation of these new modes of endocrine disruption will be key in understanding the nature of xenobiotic effects on the endocrine system.


Subject(s)
Endocrine Disruptors/toxicity , Endocrine System/drug effects , Animals , DNA Methylation/drug effects , Endocrine System/physiology , Fertility/drug effects , Hormones/metabolism , Humans , Male , Obesity/chemically induced , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Species Specificity , Spermatozoa/drug effects , Steroids/metabolism
8.
J Clin Immunol ; 24(6): 623-36, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15622447

ABSTRACT

St. John's wort is widely used as an herbal antidepressant and is among the top-selling botanical products in the United States. Although St. John's wort has been reported to have minimal side effects compared with other antidepressants, here we show that hyperforin, the active component of St. John's wort, can stimulate interleukin-8 (IL-8) expression in human intestinal epithelia cells (IEC) and primary hepatocytes. Hyperforin is also able to induce expression of mRNA, encoding another major inflammatory mediator--intercellular adhesion molecule-1 (ICAM-1). IEC participate in the intestinal inflammatory process and serve as a first line of defense through bidirectional communication between host and infectious pathogens. Although hyperforin is a potent ligand for the steroid and xenobiotic receptor (SXR), we found that hyperforin induced IL-8 mRNA through an SXR-independent transcriptional activation pathway. IL-8 induction by hyperforin required the activation of AP-1 but not the NF-kappaB transcription factor, thereby distinguishing it from the NF-kappaB-dependent IL-8 induction mediated by tumor necrosis factor alpha (TNFalpha). Further study revealed that extracellular signal-regulated kinase 1 and 2 (ERK1/2) were required for the hyperforin-induced expression of IL-8. Our results suggest a previously unsuspected effect of St. John's wort in modulating the immune and inflammatory responses.


Subject(s)
Bridged Bicyclo Compounds/pharmacology , Gene Expression Regulation/drug effects , Hypericum/chemistry , Interleukin-8/genetics , Intestinal Mucosa/drug effects , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Signal Transduction , Terpenes/pharmacology , Antidepressive Agents/pharmacology , Cell Line , Humans , Inflammation Mediators , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinases , NF-kappa B/metabolism , Phytotherapy/adverse effects , Transcription Factor AP-1/metabolism
9.
Drug Metab Dispos ; 32(10): 1075-82, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15269186

ABSTRACT

Vitamin E is an essential nutrient with antioxidant activity. Vitamin E is comprised of eight members, alpha-, beta-, gamma-, and delta-tocopherols and alpha-, beta-, gamma-, and delta-tocotrienols. All forms of vitamin E are initially metabolized by omega-oxidation, which is catalyzed by cytochrome P450 enzymes. The steroid and xenobiotic receptor (SXR) is a nuclear receptor that regulates drug clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. We show here that all four tocotrienols specifically bind to and activate SXR, whereas tocopherols neither bind nor activate. Surprisingly, tocotrienols show tissue-specific induction of SXR target genes, particularly CYP3A4. Tocotrienols up-regulate expression of CYP3A4 but not UDP-glucuronosyltransferase 1A1 (UGT1A1) or multidrug resistance protein-1 (MDR1) in primary hepatocytes. In contrast, tocotrienols induce MDR1 and UGT1A1 but not CYP3A4 expression in intestinal LS180 cells. We found that nuclear receptor corepressor (NCoR) is expressed at relatively high levels in intestinal LS180 cells compared with primary hepatocytes. The unliganded SXR interacts with NCoR, and this interaction is only partially disrupted by tocotrienols. Expression of a dominant-negative NCoR enhanced the ability of tocotrienols to induce CYP3A4 in LS180 cells, suggesting that NCoR plays an important role in tissue-specific gene regulation by SXR. Our findings provide a molecular mechanism explaining how vitamin supplements affect the absorption and effectiveness of drugs. Knowledge of drug-nutrient interactions may help reduce the incidence of decreased drug efficacy.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Gene Targeting/methods , Receptors, Steroid/metabolism , Tocotrienols/pharmacology , Cell Line , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Pregnane X Receptor , Protein Binding/drug effects , Protein Binding/physiology , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Tocotrienols/metabolism
10.
Environ Health Perspect ; 112(2): 163-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14754570

ABSTRACT

Polychlorinated biphenyls (PCBs) are a family of persistent organic contaminants suspected to cause adverse effects in wildlife and humans. In rodents, PCBs bind to the aryl hydrocarbon (AhR) and pregnane X receptors (PXR) inducing the expression of catabolic cytochrome p450 enzymes of the CYP1A and 3A families. We found that certain highly chlorinated PCBs are potent activators of rodent PXR but antagonize its human ortholog, the steroid and xenobiotic receptor (SXR), inhibiting target gene induction. Thus, exposure to PCBs may blunt the human xenobiotic response, inhibiting the detoxification of steroids, bioactive dietary compounds, and xenobiotics normally mediated by SXR. The antagonistic PCBs are among the most stable and abundant in human tissues. These findings have important implications for understanding the biologic effects of PCB exposure and the use of animal models to predict the attendant risk.


Subject(s)
Environmental Pollutants/pharmacology , Environmental Pollutants/poisoning , Polychlorinated Biphenyls/pharmacology , Polychlorinated Biphenyls/poisoning , Receptors, Steroid/drug effects , Receptors, Steroid/physiology , Xenobiotics/metabolism , Animals , Cell Culture Techniques , Diet , Humans , Inactivation, Metabolic , Mice , Pregnane X Receptor , Rats , Risk Assessment , Steroids/metabolism
11.
J Biol Chem ; 278(45): 43919-27, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-12920130

ABSTRACT

Vitamin K2 is a critical nutrient required for blood clotting that also plays an important role in bone formation. Vitamin K2 supplementation up-regulates the expression of bone markers, increases bone density in vivo, and is used clinically in the management of osteoporosis. The mechanism of vitamin K2 action in bone formation was thought to involve its normal role as an essential cofactor for gamma-carboxylation of bone matrix proteins. However, there is evidence that suggests vitamin K2 also has a transcriptional regulatory function. Vitamin K2 bound to and activated the orphan nuclear receptor SXR and induced expression of the SXR target gene, CYP3A4, identifying it as a bona fide SXR ligand. Vitamin K2 treatment of osteosarcoma cells increased mRNA levels for the osteoblast markers bone alkaline phosphatase, osteoprotegerin, osteopontin, and matrix Gla protein. The known SXR activators rifampicin and hyperforin induced this panel of bone markers to an extent similar to vitamin K2. Vitamin K2 was able to induce bone markers in primary osteocytes isolated from wild-type murine calvaria but not in cells isolated from mice deficient in the SXR ortholog PXR. We infer that vitamin K2 is a transcriptional regulator of bone-specific genes that acts through SXR to favor the expression of osteoblastic markers. Thus, SXR has a novel role as a mediator of bone homeostasis in addition to its role as a xenobiotic sensor. An important implication of this work is that a subset of SXR activators may function as effective therapeutic agents for the management of osteoporosis.


Subject(s)
Bone and Bones/metabolism , Homeostasis/drug effects , Receptors, Steroid/physiology , Vitamin K 2/pharmacology , Alkaline Phosphatase/analysis , Alkaline Phosphatase/genetics , Animals , Biomarkers/analysis , Bone Density/drug effects , Bridged Bicyclo Compounds , COS Cells , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Gene Expression/drug effects , Glycoproteins/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Osteoblasts/chemistry , Osteocalcin/genetics , Osteopontin , Osteoprotegerin , Osteosarcoma/chemistry , Phloroglucinol/analogs & derivatives , Pregnane X Receptor , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Tumor Necrosis Factor , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Rifampin/pharmacology , Sialoglycoproteins/genetics , Terpenes/pharmacology , Transfection , Tumor Cells, Cultured , Vitamin K 2/metabolism
12.
J Biol Chem ; 277(46): 43691-7, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12198127

ABSTRACT

The Xenopus benzoate nuclear hormone receptors, BXRalpha and BXRbeta, share 82% identity within their ligand-binding domains and are classified as members of the NR1I2 subfamily that includes the mammalian steroid and xenobiotic receptor, SXR/PXR. Although alkyl benzoates have been identified as endogenous ligands, the exact role of the benzoate receptors in amphibian physiology has not been established. In this report, we show that BXRalpha and BXRbeta are pharmacologically distinct from each other: BXRalpha is more promiscuous than BXRbeta with respect to both ligand specificity and co-activator recruitment. BXRalpha can be transactivated by a number of benzoate derivatives including 4-amino-butylbenzoate (4-ABB), 4-hydroxy-butylbenzoate (4-HBB), 3-hydroxy ethyl benzoate (3-HEB), and benzyl benzoate, but only 4-HBB acts as an agonist for both receptors. Furthermore, BXRalpha-specific agonists such as 4-ABB, chlorpyrifos, and trifluralin act as antagonists on BXRbeta. BXRs are widely distributed in adult tissues but do not show any enrichment in liver and intestine, major sites of SXR/PXR expression that are critical in xenobiotic metabolism. Neither BXR shows the broad specificity toward steroids or xenobiotics exhibited by SXR/PXR. Therefore, we conclude that the BXRs are pharmacologically distinct from each other and unlikely to serve as xenobiotic sensors.


Subject(s)
Benzocaine/analogs & derivatives , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Xenopus Proteins , Animals , Benzoates/pharmacology , Benzocaine/pharmacology , COS Cells , Chlorpyrifos/pharmacology , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Female , Intestinal Mucosa/metabolism , Ligands , Liver/metabolism , Male , Parabens/pharmacology , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Tissue Distribution , Transfection , Trifluralin/pharmacology , Two-Hybrid System Techniques , Xenobiotics/pharmacology , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...