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










Publication year range
1.
J Med Food ; 19(1): 47-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26623679

ABSTRACT

Phytonutrients have rapidly emerged as natural food chemicals possessing multifaceted biological actions that may support beneficial health outcomes. Among the vast array of phytonutrients currently being studied, sulforaphane, curcumin, quercetin, and resveratrol have been frequently reported to stimulate the expression of endogenous detoxification enzymes and may thereby facilitate the neutralization of otherwise harmful environmental agents. Some of these same phytonutrients, however, have also been implicated in disrupting normal cell proliferation and hence may possess toxic properties in and of themselves. In this study, we characterize the respective minimum threshold concentrations of the aforementioned phytonutrients in Hepa1c1c7 cells that stimulate NAD(P)H: quinone oxidoreductase (NQO1), a key enzyme in the hepatic neutralization of menadione, other biological oxidants, and some environmental carcinogens. Moreover, our findings demonstrate that relatively low concentrations of either sulforaphane or curcumin significantly (P < .05) increase NQO1 protein expression and activity without triggering G2/M cell cycle arrest or mitotic catastrophe. The minimal quercetin concentration inducing NQO1, however, was 100-fold higher than that which disrupted mitosis. Also, while resveratrol modestly stimulated NQO1, the minimally effective resveratrol concentration concomitantly induced evidence of cellular apoptosis. Taken together, these findings indicate that only particular phytonutrients are likely efficacious in upregulating NQO1 activity without also leading to hepatic cytotoxicity.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , Mitosis/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phytochemicals/pharmacology , Animals , Cell Line , Hepatocytes/cytology , Hepatocytes/enzymology , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics
2.
BMC Health Serv Res ; 15: 454, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26438342

ABSTRACT

BACKGROUND: Worldwide, rural communities face barriers when accessing health services. In response, numerous initiatives have focused on fostering technological innovations, new management approaches and health policies. Research suggests that the most successful innovations are those involving stakeholders at all levels. However, there is little evidence exploring the opinions of local health providers that could contribute with further innovation development and research. The aims of this study were to explore the perspectives of medical doctors (MDs) working in rural areas of Peru, regarding the barriers impacting the diagnostic process, and ideas for diagnostic innovations that could assist them. METHODS: Data gathered through three focus group discussions (FGG) and 18 individual semi-structured interviews (SSI) with MDs who had completed their medical service in rural areas of Peru in the last two years were analyzed using thematic analysis. RESULTS: Three types of barriers emerged. The first barrier was the limited access to point of care (POC) diagnostic tools. Tests were needed for: i) the differential diagnosis of malaria vs. pneumonia, ii) dengue vs. leptospirosis, iii) tuberculosis, iv) vaginal infections and cervical cancer, v) neurocysticercosis, and vi) heavy metal toxicity. Ultrasound was needed for the diagnosis of obstetric and intra-abdominal conditions. There were also health system-related barriers such as limited funding for diagnostic services, shortage of specialists, limited laboratory services and access to telecommunications, and lack of institutional support. Finally, the third type of barriers included patient related-barriers to follow through with diagnostic referrals. Ideas for innovations proposed included POC equipment and tests, and telemedicine. CONCLUSIONS: MDs at primary health facilities in rural Peru face diagnostic challenges that are difficult to overcome due to a limited access to diagnostic tools. Referrals to specialized facilities are constrained by deficiencies in the organization of health services and by barriers that impede the patients' travel to distant health facilities. Technological innovations suggested by the participants such as POC diagnostic tools and mobile-health (m-health) applications could help address part of the problem. However, other types of innovation to address social, adaptation and policy issues should not be dismissed.


Subject(s)
Health Knowledge, Attitudes, Practice , Health Services Accessibility , Medical Staff, Hospital/psychology , Rural Population , Adult , Female , Focus Groups , Humans , Male , Peru , Physicians , Pregnancy , Qualitative Research , Referral and Consultation , Telemedicine
3.
Theranostics ; 4(3): 267-79, 2014.
Article in English | MEDLINE | ID: mdl-24505235

ABSTRACT

Subtype-targeted therapies can have a dramatic impact on improving the quality and quantity of life for women suffering from breast cancer. Despite an initial therapeutic response, cancer recurrence and acquired drug-resistance are commonplace. Non-invasive imaging probes that identify drug-resistant lesions are urgently needed to aid in the development of novel drugs and the effective utilization of established therapies for breast cancer. The protease receptor urokinase plasminogen activator receptor (uPAR) is a target that can be exploited for non-invasive imaging. The expression of uPAR has been associated with phenotypically aggressive breast cancer and acquired drug-resistance. Acquired drug-resistance was modeled in cell lines from two different breast cancer subtypes, the uPAR negative luminal A subtype and the uPAR positive triple negative subtype cell line MDA-MB-231. MCF-7 cells, cultured to be resistant to tamoxifen (MCF-7 TamR), were found to significantly over-express uPAR compared to the parental cell line. uPAR expression was maintained when resistance was modeled in triple-negative breast cancer by generating doxorubicin and paclitaxel resistant MDA-MB-231 cells (MDA-MB-231 DoxR and MDA-MB-231 TaxR). Using the antagonistic uPAR antibody 2G10, uPAR was imaged in vivo by near-infrared (NIR) optical imaging and (111)In-single photon emission computed tomography (SPECT). Tumor uptake of the (111)In-SPECT probe was high in the three drug-resistant xenografts (> 46 %ID/g) and minimal in uPAR negative xenografts at 72 hours post-injection. This preclinical study demonstrates that uPAR can be targeted for imaging breast cancer models of acquired resistance leading to potential clinical applications.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Antibodies/analysis , Antineoplastic Agents/pharmacology , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Female , Humans , Immunoglobulin G/analysis , Indium Radioisotopes , MCF-7 Cells , Mice , Multimodal Imaging , Optical Imaging , Paclitaxel/pharmacology , Receptors, Urokinase Plasminogen Activator/immunology , Tamoxifen/pharmacology , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
4.
Food Chem Toxicol ; 60: 431-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939039

ABSTRACT

Curcumin, a component of turmeric spice that imparts flavor and color to curry, is thought to possess anti-inflammatory and antioxidant properties in biological tissues. However, while such efficacies have been described in the context of carcinogenesis, the impact of curcumin on normal cell cycle regulation is poorly understood. Here, we provide evidence of curcumin toxicity in proliferating bovine aortic endothelial cells, at concentrations relevant to the diet and below those previously reported in cancer models. Upon confirming curcumin's ability to upregulate hemeoxygenase-1 in a dose-dependent fashion, we found the minimally efficacious curcumin concentration to also inhibit endothelial cell DNA synthesis. Moreover, curcumin concentrations below the minimum 2 µM threshold required to induce hemeoxygenase-1 bound tubulin protein in vitro and triggered hallmark evidence of mitotic catastrophe in vivo. Concentrations as low as 0.1 µM curcumin led to disproportionate DNA segregation, karyorrhexis, and micronucleation in proliferating endothelial cells. While suggesting a mechanism by which physiological curcumin concentrations inhibit cell cycle progression, these findings describe heretofore unappreciated curcumin toxicity with potential implications for endothelial growth, development, and tissue healing.


Subject(s)
Cell Proliferation/drug effects , Curcumin/pharmacology , Endothelial Cells/drug effects , Mitosis/drug effects , Tubulin/metabolism , Animals , Antioxidants/pharmacology , Cattle , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line , Endothelial Cells/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Microtubules/drug effects , Microtubules/metabolism
5.
J Ethnopharmacol ; 139(2): 414-21, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22138349

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ginseng is a widely consumed aromatic herb that is purported to have health benefits. Several studies report a beneficial impact of ginseng or its derivatives on Candida albicans infection in mice and suggest that its immune-modulatory properties contribute to this effect. However, these studies generally administered ginseng to experimental animals by injection, whereas people typically ingest ginseng. Furthermore, although disseminated candiasis is typically a disease of immune-impaired hosts, previous studies have generally used immune competent host species in the assessments. MATERIALS AND METHODS: We evaluated the efficacy of an ingested extract of ginseng against Candida albicans infection in DBA/2J mice, which are highly susceptible to Candida albicans infection. A ginseng extract was added to the drinking water for two days before and for the remainder of the study after intravenous inoculation of mice with Candida albicans. Mice were evaluated for morbidity, mortality, Candida albicans titers, and concentrations of inflammatory cytokines and chemokines. RESULTS: Ingestion of the ginseng extract did not significantly affect overall morbidity or mortality. However, ingestion of the extract was associated with significantly lower renal titers of Candida albicans and with significantly lower concentrations of some inflammatory cytokines in kidney and/or serum. CONCLUSIONS: Assessment of morbidity, mortality, inflammatory markers, and renal titers after spontaneous ingestion of ginseng by susceptible hosts represents a comprehensive approach to characterizations of therapeutic efficacy against infectious agents. Our findings extend previous reports of the efficacy of ginseng against Candida albicans by demonstrating significant reductions in infectious load and some markers of inflammation in susceptible mice. Our data therefore support further assessment of the immune-modulatory properties of this widely consumed herb and its components.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Candidiasis/drug therapy , Panax , Plant Extracts/pharmacology , Animals , Candida albicans/growth & development , Candidiasis/immunology , Candidiasis/microbiology , Complement System Proteins/deficiency , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/microbiology , Male , Mice , Mice, Inbred DBA , Microbial Sensitivity Tests , Plants, Medicinal , Time Factors
6.
Breast Cancer Res Treat ; 128(1): 57-68, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20665104

ABSTRACT

We previously isolated cacalol as a free radical-scavenging compound from Cacalia delphiniifolia which is a traditional Asian herbal plant and is believed to have medicinal effects on cancer. In this report, we demonstrated that cacalol has strong anti-proliferation effect on breast cancer cells and induces apoptosis by activating a pro-apoptotic pathway. We also found that a combination of cacalol and other chemotherapeutic drugs (Taxol and cyclophosphamide) synergistically induced apoptosis and partially overcame chemo-resistance. To further gain a mechanistic insight, we tested a potential inhibitory effect of cacalol on fatty acid synthase gene (FAS) in breast cancer cells, and found that cacalol significantly modulated the expression of the FAS gene, which resulted in apoptosis through activation of DAPK2 and caspase 3. We have also shown that cacalol significantly suppressed the Akt-sterol regulatory element-binding proteins (SREBP) signaling pathway and concomitant transcriptional activation of FAS. In a xenograft model of nude mouse, when cacalol was administered intraperitoneally, tumor growth was significantly suppressed. Importantly, oral administration of cacalol before implanting tumors showed significant preventive effect on tumor growth in the same animal model. Furthermore, the treatment of mice with a combination of low dose of Taxol and cacalol significantly suppressed the tumor growth. Taken together, our results indicate that cacalol induces apoptosis in breast cancer cells and impairs mammary tumor growth in vivo by blocking the expression of the FAS gene through modulation of Akt-SREBP pathway, suggesting that cacalol has potential utility as a chemopreventive and chemotherapeutic agent for breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fatty Acid Synthases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sesquiterpenes/pharmacology , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Antineoplastic Agents/adverse effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cyclophosphamide/pharmacology , Death-Associated Protein Kinases , Drug Synergism , Enzyme Activation , Fatty Acid Synthases/genetics , Female , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Mice, Nude , Paclitaxel/pharmacology , Promoter Regions, Genetic , Sesquiterpenes/adverse effects , Signal Transduction , Transcription, Genetic , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
J Surg Res ; 157(2): 261-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19815237

ABSTRACT

BACKGROUND: Epidemiologic evidence suggests reduced breast cancer mortality in users of American Ginseng (AG) (Panax quinquefolium). We hypothesized that AG extract decreases proliferation of human breast cancer cells via an anti-inflammatory effect applicable to the prevention of breast and other cancers. MATERIAL AND METHODS: A defined lyophilized aqueous extract of AG (LEAG) was dissolved in DMSO 1mg/mL, and serially diluted in saline. The cell lines MDA MB 231 and MCF7 were stimulated with the phorbol ester PDBu and treated with 100-500 mcg/mL LEAG. Proliferation was measured by MDA assay. Induced COX-2 expression was assayed by ELISA. Activation of NFkappaB by phosphorylation of the p65 subunit was quantified by CASE (cellular activation of signaling ELISA). RESULTS: Both cell lines had reduced proliferation when treated with LEAG. PDBu stimulation of MDA MB 231 increased expression of the COX-2 protein 20-fold at 48 hours (P<0.005). COX-2 protein expression remained at baseline concentrations in PDBu- treated MDA MB 231 cells exposed to 100 mcg/mL LEAG. The CASE assay showed a 4-fold increase in p65 activation 24 hours after PDBu treatment in normal medium, while phosphorylated p65 dropped below baseline in the cells treated with PDBu plus LEAG. CONCLUSION: In MDA MB 231, COX-2 was inducible with PDBu. This induced COX-2 expression was blocked by 100 microgram/mL LEAG in a time course consistent with the decline in the activated p65 subunit of NFkappaB. These results provide an anti-inflammatory mechanism for a possible anti-cancer effect of American Ginseng.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , NF-kappa B/metabolism , Panax , Plant Extracts/pharmacology , Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclin D1/metabolism , Humans
8.
J Exp Ther Oncol ; 6(2): 147-55, 2007.
Article in English | MEDLINE | ID: mdl-17407973

ABSTRACT

Ginseng has been shown to inhibit cancer cell proliferation and tumor growth, however the mechanisms underlying this inhibition have yet to be elucidated. An inhibitory effect of hot water-extracted American ginseng (Panax quinquefolius L.) root on cell proliferation was demonstrated using MCF-7 human breast cancer cells treated with a wide concentration range of the ginseng extract (GE) for 6 days. The effects of GE were concentration-dependent with an IC50 of 0.49 microg/microl and the minimum exposure time to elicit an inhibitory response was 24 hours. Using an antibody microarray, it was determined that several key cell survival proteins were altered in GE-treated cells, including several members of the mitogen-activated protein kinase (MAPK) family. A GE-induced decrease in phospho-MEK1/2 and -ERK1/2 and an increase in phospho-Raf-1 were observed and verified using Western blot analysis. Furthermore, mRNA and protein expression of the Raf-1 kinase inhibitor protein (RKIP) was shown to be transiently, yet significantly, upregulated following GE treatment. These results suggest that American ginseng may act to inhibit breast cancer cell proliferation by increasing the expression of RKIP, resulting in inhibition of the MAPK pathway. This novel mechanism has implications in the potential prevention and treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Panax , Plant Extracts/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/physiology , Phosphorylation , RNA, Messenger/analysis
9.
Integr Cancer Ther ; 5(3): 236-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16880429

ABSTRACT

HYPOTHESIS: Ginseng root extracts and the biologically active ginsenosides have been shown to inhibit proliferation of human cancer cell lines, including breast cancer. However, there are conflicting data that suggest that ginseng extracts (GEs) may or may not have estrogenic action, which might be contraindicated in individuals with estrogen-dependent cancers. The current study was designed to address the hypothesis that the extraction method of American ginseng (Panax quinquefolium) root will dictate its ability to produce an estrogenic response using the estrogen receptor (ER)-positive MCF-7 human breast cancer cell model. METHODS: MCF-7 cells were treated with a wide concentration range of either methanol-(alc-GE) or water-extracted (w-GE) ginseng root for 6 days. Cells were grown in media containing either normal or charcoal-stripped fetal calf serum to limit exposure to exogenous estrogen. Thus, an increase in MCF-7 cell proliferation by GE indicated potential estrogenicity. This was confirmed by blocking GE-induced MCF-7 cell proliferation with ER antagonists ICI 182,780 (1 nM) and 4-hydroxytamoxifen (0.1 microM). Furthermore, the ability of GE to bind ERalpha or ERbeta and stimulate estrogen-responsive genes was examined. RESULTS: Alc-GE, but not w-GE, was able to increase MCF-7 cell proliferation at low concentrations (5-100 microg/mL) when cells were maintained under low-estrogen conditions. The stimulatory effect of alc-GE on MCF-7 cell proliferation was blocked by the ER antagonists ICI 182,780 or 4-hydroxyta-moxifen. At higher concentrations of GE, both extracts inhibited MCF-7 and ER-negative MDA-MB-231 cell proliferation regardless of media conditions. Binding assays demonstrated that alc-GE, but not w-GE, was able to bind ERalpha and ERbeta. Alc-GE (50 microg/mL) also induced an approximate 2.5-fold increase in expression of the estrogen-responsive pS2 gene, as well as progesterone receptor (PgR) gene expression, whereas w-GE was without effect. CONCLUSION: These data indicate that low concentrations of alc-GE, but not w-GE, elicit estrogenic effects, as evidenced by increased MCF-7 cell proliferation, in a manner antagonized by ER antagonists, interactions of alc-GE with estrogen receptors, and increased expression of estrogen-responsive genes by alc-GE. Thus, discrepant results between different laboratories may be due to the type of GE being analyzed for estrogenic activity.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chemistry, Pharmaceutical/methods , Panax , Receptors, Estrogen/drug effects , Saponins/pharmacology , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Line, Tumor , Ethanol , Humans , Plant Extracts/pharmacology , Plant Roots , Receptors, Estrogen/metabolism , Water
10.
Life Sci ; 78(12): 1336-40, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16288926

ABSTRACT

American ginseng root (Panax quinquefolius) has a number of purported therapeutic effects, including inhibition of cancer cell proliferation. The ability of environmentally relevant heavy metals to alter ginseng effects on cancer cell growth was the subject of this study. A water extract of American ginseng root was applied alone or in combination with physiologically relevant doses of either lead (Pb) or arsenite to MCF-7 breast cancer cells in vitro and effects on cell proliferation were determined. Ginseng alone produced a significant dose-dependent inhibition of MCF-7 cell proliferation starting at 0.5 mg ml(-1). Treatment of MCF-7 cells with 2.5 microM arsenite significantly decreased MCF-7 cell proliferation (p < 0.01). When cells were treated with arsenite (1.25 or 2.5 microM) in combination with ginseng extract (0.5 mg ml(-1)), there was an apparent synergistic inhibition of cell proliferation. Treatment of MCF-7 breast cancer cells with 50 microM Pb significantly decreased cell proliferation relative to control (p < 0.01), and concomitant ginseng and Pb treatment did not lead to a further decrease. These results suggest that contaminant heavy metals, some of which have been detected in ginseng root extracts or commercial ginseng preparations, may alter the biological activity of ginseng.


Subject(s)
Arsenites/therapeutic use , Cell Division/drug effects , Lead/pharmacology , Panax , Plant Extracts/pharmacology , Plant Roots , Breast Neoplasms , Cell Line, Tumor , Female , Humans , Phytotherapy
11.
J Agric Food Chem ; 53(26): 9867-73, 2005 Dec 28.
Article in English | MEDLINE | ID: mdl-16366667

ABSTRACT

Four methods were tested for extraction and recovery of six major ginsenosides (Rb1, Rb2, Rc, Rd, Re, and Rg1) found in roots of American ginseng (Panax quinquefolius): method A, sonication in 100% methanol (MeOH) at room temperature (rt); method B, sonication in 70% aqueous MeOH at rt; method C, water extraction (90 degrees C) with gentle agitation; and method D, refluxing (60 degrees C) in 100% MeOH. After 0.5-1 h, the samples were filtered and analyzed by high-performance liquid chromatography (HPLC)-UV. A second extraction by methods C and D was done, but 85-90% of ginsenosides were obtained during the first extraction. Lyophilization of extracts did not influence ginsenoside recovery. Method D resulted in the highest significant recoveries of all ginsenosides, except Rg1. Method C was the next most effective method, while method A resulted in the lowest ginsenoside recoveries. Method B led to similar recoveries as method C. All methods used one filtration step, omitted time-consuming cleanup, but maintained clear peak resolution by HPLC, and can be used for quantitative screening of ginsenosides from roots and commercial ginseng preparations.


Subject(s)
Ginsenosides/isolation & purification , Panax/chemistry , Chromatography, High Pressure Liquid , Methanol , Plant Extracts/analysis , Plant Roots/chemistry , Reference Standards , Solvents , Spectrophotometry, Ultraviolet , Water
12.
Ann N Y Acad Sci ; 962: 372-7, 2002 May.
Article in English | MEDLINE | ID: mdl-12076988

ABSTRACT

In Asia, ginseng is commonly included in herbals used for the treatment of sexual dysfunction. Recent studies in laboratory animals have shown that both Asian and American forms of ginseng enhance libido and copulatory performance. These effects of ginseng may not be due to changes in hormone secretion, but to direct effects of ginseng, or its ginsenoside components, on the central nervous system and gonadal tissues. Indeed, there is good evidence that ginsenosides can facilitate penile erection by directly inducing the vasodilatation and relaxation of penile corpus cavernosum. Moreover, the effects of ginseng on the corpus cavernosum appear to be mediated by the release and/or modification of release of nitric oxide from endothelial cells and perivascular nerves. Treatment with American ginseng also affects the central nervous system and has been shown to significantly alter the activity of hypothalamic catecholamines involved in the facilitation of copulatory behavior and hormone secretion. Recent findings that ginseng treatment decreased prolactin secretion also suggested a direct nitric oxide-mediated effect of ginseng at the level of the anterior pituitary. Thus, animal studies lend growing support for the use of ginseng in the treatment of sexual dysfunction and provide increasing evidence for a role of nitric oxide in the mechanism of ginsenoside action.


Subject(s)
Aphrodisiacs , Nitric Oxide/metabolism , Panax , Sexual Behavior , Animals , Drugs, Chinese Herbal/metabolism , Hormones/metabolism , Humans , Panax/chemistry , Panax/metabolism , Sexual Behavior, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...