Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Birth Defects Res ; 111(2): 70-77, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30575315

ABSTRACT

BACKGROUND: Development of an in vitro system capable of producing mature sperm remains a challenging goal, with only few successes reported. Such a system, could be used to test agents for potential toxicity to the male reproductive system; to explore this, we exposed immature mouse testis fragments in culture to ethinylestradiol (EE), a well-known testicular toxicant in vivo. METHODS: Testis fragments from postnatal day 5 mice were cultured in Albumax I medium. After 24 hr of culture, fragments were treated with 0.01, 0.1 or 1 nM EE, then harvested after 20 days in culture and examined for histology or gene expression measures by quantitative PCR. RESULTS: There was substantial variability between fragments in the degree of spermatogenesis observed. The percentage of seminiferous tubules containing any dead germ cells increased as a result of EE exposure in a dose dependent fashion. This was accompanied with a decreased percentage of tubules with round spermatids. Expression of estrogen receptor 1, cytochrome P450, family 11, subfamily a, and polypeptide 1 also was reduced, depending on the dose. CONCLUSION: These gene expression changes in the testis fragments are similar to those seen after animals have been exposed to EE. Gene expression changes in testis fragments are encouraging, but the variability across samples will need to be reduced for this in vitro system to become a generally applicable method for assessing testicular toxicants.


Subject(s)
Organ Culture Techniques/methods , Spermatogenesis/physiology , Tissue Culture Techniques/methods , Animals , Cell Differentiation , Ethinyl Estradiol/pharmacology , Genitalia, Male/cytology , Male , Mice , Mice, Inbred C57BL , Seminiferous Tubules/metabolism , Spermatids/drug effects , Spermatogenesis/drug effects , Spermatozoa/drug effects , Testis/metabolism
2.
J Toxicol Sci ; 44(10): 667-679, 2019.
Article in English | MEDLINE | ID: mdl-31588058

ABSTRACT

The assessment of xenobiotic-induced testicular toxicity is important in drug development. Nonetheless, in vitro models to test drugs and chemicals that may cause testicular toxicity are lacking, requiring the continued use of animal models for those studies. We previously evaluated an in vitro mouse testis organ culture system using ethinylestradiol (EE), a well-studied testicular toxicant, and demonstrated a dose-dependent relationship between adverse effects to germ cell differentiation and increasing EE concentrations. However, we terminated that study after 20 days of culture due to oxygen deficiency during germ cell differentiation. Therefore, in the current study, we aimed to identify gene(s) with potential for supporting the histopathological evaluations of testicular toxicity using in vitro testis organ culture system. We cultured testis fragments obtained from mice at postnatal day (PND) 5 in α-Minimal Essential Medium containing 40 mg/mL AlbuMAX™ I and treated them with 0.01 or 1 nM EE on day 1 of culture. On day 20, we collected testis fragments for RNA sequencing analysis and quantitative polymerase chain reaction (qPCR). We found that phospholipase C, zeta 1 and testis-specific serine kinase 4 genes, that are involved in spermatogenesis and predominantly expressed in the testis, were significantly reduced in testis fragments treated with the highest concentration of EE. Also, cytochrome P450, family 26, subfamily b, polypeptide 1 (Cyp26b1) and interleukin 16 (Il16) were up-regulated in the highest EE-treated groups. Further studies are needed to confirm the variations of these gene expression using other testicular toxicants.


Subject(s)
Estrogens/toxicity , Ethinyl Estradiol/toxicity , Testis/drug effects , Transcriptome/drug effects , Animals , Gene Expression Profiling , Male , Mice, Inbred C57BL , Organ Culture Techniques , Polymerase Chain Reaction , Sequence Analysis, RNA , Testis/metabolism , Testis/pathology
3.
Birth Defects Res ; 111(5): 270-280, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30703285

ABSTRACT

BACKGROUND: Previously, we evaluated optimal organ culture conditions to produce elongated spermatids in an in vitro mouse testis culture system. However, differences in testicular function between the cultured testis fragments and animal testis have not been determined. METHODS: To examine how closely cultured testis fragments in vitro approximates what typically occurs during the first wave of spermatogenesis in vivo, C57BL/6J mouse testis fragments obtained on postnatal day (PND) 5 were cultured in AlbuMAX™ I/ α-Minimal Essential Medium for 15, 23, 30, 35, 42, and 49 days, and compared to mouse testes obtained at PND 5, 14, 20, 24, 28, 30, 35, and 40. At the specified days of culture or PND of mice, the following analyses were conducted: histology, flow cytometry for haploid cell detection, qPCR for spermatid markers, and liquid chromatography/mass spectrometry for testosterone levels. RESULTS: Round spermatids were initially observed at 23 days, and their percentage of the total number of cells continued to increase with culture time, as did gene expression of the spermatid markers and haploid cell percentage in the cultured testis fragments. These results were similar in temporal sequence to those in animals. Testosterone levels in the testis fragments reached a maximum at Day 49. CONCLUSION: These findings show this in vitro mouse testis organ culture model may be a useful and convenient tool for mechanistic studies. However, because germ cell differentiation in all seminiferous tubules was not observed, improvements in the system/methods are needed to more closely replicate spermatogenesis as observed in animals.


Subject(s)
Organ Culture Techniques/methods , Spermatogenesis/physiology , Testis/metabolism , Animals , Cell Differentiation , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture/methods , Seminiferous Tubules/metabolism , Spermatids/cytology , Spermatozoa/cytology , Testis/physiology
4.
Clin Cancer Res ; 25(20): 6021-6025, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31064779

ABSTRACT

On November 21, 2018, the FDA approved glasdegib (Daurismo; Pfizer), a small-molecule Hedgehog inhibitor, in combination with low-dose cytarabine (LDAC) for treatment of newly diagnosed acute myeloid leukemia (AML) in adults ≥ 75 years or with comorbidities that preclude use of intensive induction chemotherapy. Evidence of clinical benefit came from Study BRIGHT AML 1003, a randomized trial comparing glasdegib+LDAC with LDAC alone for treatment of newly diagnosed AML in 115 patients either ≥ 75 years old or ≥ 55 years old with preexisting comorbidities. Efficacy was established by improved overall survival (OS) with the combination compared with LDAC alone (HR, 0.46; 95% confidence interval, 0.30-0.71; one-sided stratified log-rank P = 0.0002). Median OS was 8.3 months with the combination and 4.3 months with LDAC alone. Common adverse reactions included cytopenias, fatigue, hemorrhage, febrile neutropenia, musculoskeletal pain, nausea, edema, dyspnea, decreased appetite, dysgeusia, mucositis, constipation, and rash. The label includes a boxed warning for embryo-fetal toxicity and a warning for QT interval prolongation. There is a limitation of use for patients with moderate-to-severe hepatic and severe renal impairment; trials studying glasdegib in these patient populations are required as a condition of this approval.See related commentary by Fathi, p. 6015.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , Cytarabine/administration & dosage , Drug Approval , Leukemia, Myeloid, Acute/drug therapy , Phenylurea Compounds/administration & dosage , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Comorbidity , Cytarabine/adverse effects , Dose-Response Relationship, Drug , Drug Labeling , Female , Humans , Induction Chemotherapy/methods , Leukemia, Myeloid, Acute/epidemiology , Male , Middle Aged , Phenylurea Compounds/adverse effects , Smoothened Receptor/antagonists & inhibitors , Survival Analysis , Treatment Outcome , United States , United States Food and Drug Administration
5.
Clin Cancer Res ; 24(14): 3242-3246, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29476018

ABSTRACT

On September 1, 2017, the FDA granted approval for gemtuzumab ozogamicin (Mylotarg; Pfizer Inc.) in combination with daunorubicin and cytarabine and as a monotherapy for the treatment of adult patients with newly diagnosed CD33-positive acute myeloid leukemia (AML). Gemtuzumab ozogamicin is a CD33-targeted antibody-drug conjugate joined to calicheamicin. Approval of gemtuzumab ozogamicin combination treatment was based on a randomized trial of 271 patients with newly diagnosed AML treated with daunorubicin and cytarabine with or without 3 mg/m2 fractionated gemtuzumab ozogamicin, which resulted in an event-free survival (EFS) of 13.6 months for gemtuzumab ozogamicin + daunorubicin and cytarabine and 8.8 months for daunorubicin and cytarabine alone [HR = 0.68 (95% confidence interval (CI), 0.51-0.91)]. Hemorrhage, prolonged thrombocytopenia, and veno-occlusive disease were serious toxicities that were more common in patients treated with gemtuzumab ozogamicin + daunorubicin and cytarabine. Approval of gemtuzumab ozogamicin monotherapy was based on a randomized trial of 237 patients with newly diagnosed AML treated without curative intent. Median overall survival (OS) was 4.9 months with gemtuzumab ozogamicin versus 3.6 months on best supportive care [HR = 0.69 (95% CI, 0.53-0.90)]. Adverse events were similar on both arms. Postapproval, several studies are required including evaluation of fractionated gemtuzumab ozogamicin pharmacokinetics, safety of combination gemtuzumab ozogamicin in the pediatric population, immunogenicity, and the effects of gemtuzumab ozogamicin on platelet function. Clin Cancer Res; 24(14); 3242-6. ©2018 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Drug Approval , Gemtuzumab/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Adult , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gemtuzumab/pharmacology , Humans , Leukemia, Myeloid, Acute/diagnosis , Treatment Outcome
6.
Birth Defects Res ; 109(7): 465-474, 2017 04 17.
Article in English | MEDLINE | ID: mdl-28398669

ABSTRACT

BACKGROUND: The complexity of spermatogenesis makes development of appropriate in vitro testis models challenging. A novel in vitro mouse testis culture system has been reported but not yet evaluated as an alternative model for male reproductive toxicity testing. We assessed the effects of media composition on sperm differentiation and testis morphology of cultured mouse testis fragments. METHODS: Testes from postnatal day 5 B6:CBA-Tg(Acrv1-EGFP)2727Redd/J male mice were cultured in knockout serum replacement (KSR) or Albumax I (Albumax) medium. Enhanced green fluorescent protein (EGFP) expression was examined on days 35, 42, 45, and 49 of culture. Histology and flow cytometry were performed for testis morphology and spermatid differentiation. RESULTS: EGFP signals were first observed in round spermatids on day 22 of culture (corresponding to postnatal day 27) and were observed until the end of culture, indicating testis-specific protein expression. A-kinase anchor protein 4 expression, a marker of elongated spermatid (step 15-16) occurred earlier in explants cultured in KSR than Albumax medium (typically day 35 and after day 42 of culture, respectively). The percentage of seminiferous tubules with elongated spermatid was higher in Albumax than KSR medium from days 45 to 49 of culture. CONCLUSION: Albumax medium may facilitate or support better morphology and spermatid production than KSR medium. Further studies need to improve spermatid production and refinement of this in vitro testis culture system that may be useful as a supplement to current male reproductive toxicity testing or an alternative model in cases where in vivo testing may be unfeasible. Birth Defects Research 109:465-474, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Organ Culture Techniques/methods , Testis/physiology , Animals , Cells, Cultured , Culture Media/metabolism , Genitalia, Male/physiology , Green Fluorescent Proteins , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Models, Animal , Seminiferous Tubules/metabolism , Serum/metabolism , Spermatids/cytology , Spermatogenesis/physiology , Testis/metabolism , Toxicity Tests
7.
Clin Cancer Res ; 21(12): 2666-70, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25802282

ABSTRACT

On July 3, 2014, the FDA granted accelerated approval for belinostat (Beleodaq; Spectrum Pharmaceuticals, Inc.), a histone deacetylase inhibitor, for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma (PTCL). A single-arm, open-label, multicenter, international trial in the indicated patient population was submitted in support of the application. Belinostat was administered intravenously at a dose of 1000 mg/m(2) over 30 minutes once daily on days 1 to 5 of a 21-day cycle. The primary efficacy endpoint was overall response rate (ORR) based on central radiology readings by an independent review committee. The ORR was 25.8% [95% confidence interval (CI), 18.3-34.6] in 120 patients that had confirmed diagnoses of PTCL by the Central Pathology Review Group. The complete and partial response rates were 10.8% (95% CI, 5.9-17.8) and 15.0% (95% CI, 9.1-22.7), respectively. The median duration of response, the key secondary efficacy endpoint, was 8.4 months (95% CI, 4.5-29.4). The most common adverse reactions (>25%) were nausea, fatigue, pyrexia, anemia, and vomiting. Grade 3/4 toxicities (≥5.0%) included anemia, thrombocytopenia, dyspnea, neutropenia, fatigue, and pneumonia. Belinostat is the third drug to receive accelerated approval for the treatment of relapsed or refractory PTCL.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Approval , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/pathology , Sulfonamides/therapeutic use , United States Food and Drug Administration , Humans , United States
8.
In Vitro Cell Dev Biol Anim ; 38(4): 218-27, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12197774

ABSTRACT

Rabbit kidney proximal convoluted tubule (RPCT) and proximal straight tubule (RPST) cells were independently isolated and cultured. The kinetics of the sodium-dependent glucose transport was characterized by determining the uptake of the glucose analog alpha-methylglucopyranoside. Cell culture and assay conditions used in these experiments were based on previous experiments conducted on the renal cell line derived from the whole kidney of the Yorkshire pig (LLC-PK1). Results indicated the presence of two distinct sodium-dependent glucose transporters in rabbit renal cells: a relatively high-capacity, low-affinity transporter (V(max) = 2.28 +/- 0.099 nmoles/mg protein min, Km = 4.1 +/- 0.27 mM) in RPCT cells and a low-capacity, high-affinity transporter (V(max) = 0.45 +/- 0.076 nmoles/mg protein min, K(m) = 1.7 +/- 0.43 mM) in RPST cells. A relatively high-capacity, low-affinity transporter (V(max) = 1.68 +/- 0.215 nmoles/mg protein min, Km = 4.9 +/- 0.23 mM) was characterized in LLC-PK1 cells. Phlorizin inhibited the uptake of alpha-methylglucopyranoside in proximal convoluted, proximal straight, and LLC-PK1 cells by 90, 50, and 90%, respectively. Sodium-dependent glucose transport in all three cell types was specific for hexoses. These data are consistent with the kinetic heterogeneity of sodium-dependent glucose transport in the S1-S2 and S3 segments of the mammalian renal proximal tubule. The RPCT-RPST cultured cell model is novel, and this is the first report of sodium-dependent glucose transport characterization in primary cultures of proximal straight tubule cells. Our results support the use of cultured monolayers of RPCT and RPST cells as a model system to evaluate segment-specific differences in these renal cell types.


Subject(s)
Glucose/metabolism , Kidney Tubules, Proximal/cytology , Monosaccharide Transport Proteins/metabolism , Animals , Biological Transport , Cell Culture Techniques/methods , Cell Line , Cells, Cultured , Kidney Tubules, Proximal/physiology , Kinetics , Rabbits , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL