Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cuad Bioet ; 34(111): 175-188, 2023.
Article in Spanish | MEDLINE | ID: mdl-37804491

ABSTRACT

Human-animal chimera research has gradually evolved to the present day, in which large projects related to the attempt to solve pathologies that help us human beings to alleviate diseases. However, it must be considered that many of these advances in science imply an important ethical dilemma in many cases, and even more so if we involve people in said experiments. In the present systematic review we sought to identify these ethical problems related to chimeras, as well as possible solutions to them proposed in the literature, including technical means for the realization of less humanized chimeras. A bibliographic search was carried out in the Pubmed, Embase and Medes databases on January 4 th, 2022. The articles that strictly comply with the objectives selected for the completion of the work will be selected. A total of 21 articles makes up our sample, from which ethical problems related to chimeras, possible solutions and technical means to avoid obtaining too humanized chimeras will be extracted. The issues identified in the articles are problems related to animal welfare, acquisition of human traits from chimeras, medical concerns derived from experimentation such as zoonoses, the origin of pluripotential cells for chimera production, the creation of human gametes by said chimeras, neurological chimerism and the moral status of chimeras. This paper provides solutions for these problems, such as the use of suicide genes in human cells that would be activated if they differentiate into neuronal cells or the use of gene editing through the CRISPR/Cas9 mechanism to incapacitate these cells so that they do not differentiate into neuronal cells. The only question that remains elusive to the proposal of solutions is the one related to the potential moral status of chimeras. It is certainly a complex issue given the variety of proposals on the concept of moral status available in literature. It is therefore necessary to bring these proposals closer to reflection on human-animal chimeras in order to initiate a discussion that can shed light on this issue.


Subject(s)
Chimera , Ethics, Research , Animals , Humans
2.
Cuad. bioét ; 34(111): 178-188, may.- ago. 2023. tab, graf
Article in Spanish | IBECS | ID: ibc-226232

ABSTRACT

Las investigaciones con quimeras humano-animales han evolucionado gradualmente hasta día de hoy, en que se plantean grandes proyectos relacionados con el intento de solucionar patologías que nos ayu den a los seres humanos a paliar enfermedades. Sin embargo, se debe de tener en cuenta, que muchos de estos avances científicos llevan implícito un dilema ético importante en muchos casos, y más si se involucra a personas en dichos experimentos. En la presente revisión sistemática se buscó identificar estos problemas éticos relacionados con las quimeras, así como posibles soluciones a los mismos propuestas en la literatura, incluyendo medios técnicos para la realización de quimeras menos humanizadas. Se realizó una búsqueda bibliográfica sistemática en las bases de datos Pubmed, Embase y Medes con fecha 4 de enero de 2022. Se seleccionan los artículos que cumplían estrictamente con los objetivos seleccionados para la realización del trabajo. Un total de 21 artículos componen nuestra muestra, de los cuales se extraen problemas éticos relacionados con las quimeras, posibles soluciones y medios técnicos para evitar la obtención de quimeras demasiado humanizadas. Las cuestiones identificadas en los artículos seleccionados son problemas relacio nados con el bienestar animal, adquisición de rasgos humanos de las quimeras, preocupaciones médicas derivadas de la experimentación como pueden ser las zoonosis, el origen de las células pluripontenciales para la realización de quimeras, la creación de gametos humanos por parte de dichas quimeras, el qui merismo neurológico y el estatus moral de las quimeras. En el trabajo se aportan soluciones para estos problemas, tales como la utilización de genes suicidas en las células humanas que se activarían si estas se diferencian en células neuronales o el uso de la edición genética mediante el mecanismo CRISPR/Cas9 para incapacitar a estas células para que no se diferencien en células neuronales (AU)


Human-animal chimera research has gradually evolved to the present day, in which large projects re lated to the attempt to solve pathologies that help us human beings to alleviate diseases. However, it must be considered that many of these advances in science imply an important ethical dilemma in many cases, and even more so if we involve people in said experiments. In the present systematic review we sought to identify these ethical problems related to chimeras, as well as possible solutions to them proposed in the literature, including technical means for the realization of less humanized chimeras. A bibliographic search was carried out in the Pubmed, Embase and Medes databases on January 4th, 2022. The articles that strictly comply with the objectives selected for the completion of the work will be selected. A total of 21 articles makes up our sample, from which ethical problems related to chimeras, possible solutions and technical means to avoid obtaining too humanized chimeras will be extracted. The issues identified in the articles are problems related to animal welfare, acquisition of human traits from chimeras, medical concerns derived from experimentation such as zoonoses, the origin of pluripotential cells for chimera production, the cre ation of human gametes by said chimeras, neurological chimerism and the moral status of chimeras. This paper provides solutions for these problems, such as the use of suicide genes in human cells that would be activated if they differentiate into neuronal cells or the use of gene editing through the CRISPR/Cas9 mech anism to incapacitate these cells so that they do not differentiate into neuronal cells. The only question that remains elusive to the proposal of solutions is the one related to the potential moral status of chime ras (AU)


Subject(s)
Humans , Animal Experimentation/ethics , Human Experimentation/ethics , Ethics, Research , Chimera
4.
Oncogene ; 40(5): 997-1011, 2021 02.
Article in English | MEDLINE | ID: mdl-33323970

ABSTRACT

Estrogen receptor alpha gene (ESR1) mutations occur frequently in ER-positive metastatic breast cancer, and confer clinical resistance to aromatase inhibitors. Expression of the ESR1 Y537S mutation induced an epithelial-mesenchymal transition (EMT) with cells exhibiting enhanced migration and invasion potential in vitro. When small subpopulations of Y537S ESR1 mutant cells were injected along with WT parental cells, tumor growth was enhanced with mutant cells becoming the predominant population in distant metastases. Y537S mutant primary xenograft tumors were resistant to the antiestrogen tamoxifen (Tam) as well as to estradiol (E2) withdrawal. Y537S ESR1 mutant primary tumors metastasized efficiently in the absence of E2; however, Tam treatment significantly inhibited metastasis to distant sites. We identified a nine-gene expression signature, which predicted clinical outcomes of ER-positive breast cancer patients, as well as breast cancer metastasis to the lung. Androgen receptor (AR) protein levels were increased in mutant models, and the AR agonist dihydrotestosterone significantly inhibited estrogen-regulated gene expression, EMT, and distant metastasis in vivo, suggesting that AR may play a role in distant metastatic progression of ESR1 mutant tumors.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , Receptors, Androgen/genetics , Tamoxifen/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estradiol/metabolism , Estrogens/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mutation/genetics , Neoplasm Metastasis , Receptors, Androgen/drug effects , Xenograft Model Antitumor Assays
5.
Br J Cancer ; 124(1): 191-206, 2021 01.
Article in English | MEDLINE | ID: mdl-33257837

ABSTRACT

BACKGROUND: Oestrogen Receptor 1 (ESR1) mutations are frequently acquired in oestrogen receptor (ER)-positive metastatic breast cancer (MBC) patients who were treated with aromatase inhibitors (AI) in the metastatic setting. Acquired ESR1 mutations are associated with poor prognosis and there is a lack of effective therapies that selectively target these cancers. METHODS: We performed a proteomic kinome analysis in ESR1 Y537S mutant cells to identify hyperactivated kinases in ESR1 mutant cells. We validated Recepteur d'Origine Nantais (RON) and PI3K hyperactivity through phospho-immunoblot analysis, organoid growth assays, and in an in vivo patient-derived xenograft (PDX) metastatic model. RESULTS: We demonstrated that RON was hyperactivated in ESR1 mutant models, and in acquired palbociclib-resistant (PalbR) models. RON and insulin-like growth factor 1 receptor (IGF-1R) interacted as shown through pharmacological and genetic inhibition and were regulated by the mutant ER as demonstrated by reduced phospho-protein expression with endocrine therapies (ET). We show that ET in combination with a RON inhibitor (RONi) decreased ex vivo organoid growth of ESR1 mutant models, and as a monotherapy in PalbR models, demonstrating its therapeutic efficacy. Significantly, ET in combination with the RONi reduced metastasis of an ESR1 Y537S mutant PDX model. CONCLUSIONS: Our results demonstrate that RON/PI3K pathway inhibition may be an effective treatment strategy in ESR1 mutant and PalbR MBC patients. Clinically our data predict that ET resistance mechanisms can also contribute to CDK4/6 inhibitor resistance.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Female , Humans , Mice , Mutation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays
6.
BMC Vet Res ; 16(1): 264, 2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32727468

ABSTRACT

BACKGROUND: Neonatal diarrhea remains one of the main causes of morbi-mortality in dairy calves under artificial rearing. It is often caused by infectious agents of viral, bacterial, or parasitic origin. Cows vaccination and colostrum intake by calves during the first 6 h of life are critical strategies to prevent severe diarrhea but these are still insufficient. Here we report the field evaluation of a product based on IgY antibodies against group A rotavirus (RVA), coronavirus (CoV), enterotoxigenic Escherichia coli, and Salmonella sp. This product, named IgY DNT, has been designed as a complementary passive immunization strategy to prevent neonatal calf diarrhea. The quality of the product depends on the titers of specific IgY antibodies to each antigen evaluated by ELISA. In the case of the viral antigens, ELISA antibody (Ab) titers are correlated with protection against infection in calves experimentally challenged with RVA and CoV (Bok M, et al., Passive immunity to control bovine coronavirus diarrhea in a dairy herd in Argentina, 2017), (Vega C, et al., Vet Immunol Immunopathol, 142:156-69, 2011), (Vega C, et al., Res Vet Sci, 103:1-10, 2015). To evaluate the efficiency in dairy farms, thirty newborn Holstein calves were randomly assigned to IgY DNT or control groups and treatment initiated after colostrum intake and gut closure. Calves in the IgY DNT group received 20 g of the oral passive treatment in 2 L of milk twice a day during the first 2 weeks of life. Animals were followed until 3 weeks of age and diarrhea due to natural exposure to infectious agents was recorded during all the experimental time. RESULTS: Results demonstrate that the oral administration of IgY DNT during the first 2 weeks of life to newborn calves caused a delay in diarrhea onset and significantly reduced its severity and duration compared with untreated calves. Animals treated with IgY DNT showed a trend towards a delay in RVA infection with significantly shorter duration and virus shedding compared to control calves. CONCLUSIONS: This indicates that IgY DNT is an effective product to complement current preventive strategies against neonatal calf diarrhea in dairy farms. Furthermore, to our knowledge, this is the only biological product available for the prevention of virus-associated neonatal calf diarrhea.


Subject(s)
Antibodies, Bacterial/therapeutic use , Antibodies, Viral/therapeutic use , Cattle Diseases/therapy , Diarrhea/veterinary , Immunoglobulins/therapeutic use , Immunotherapy , Animals , Animals, Newborn , Antibodies, Protozoan , Cattle , Cattle Diseases/microbiology , Cattle Diseases/virology , Cryptosporidiosis/prevention & control , Dairying , Diarrhea/microbiology , Diarrhea/therapy , Diarrhea/virology , Immunization, Passive/methods , Immunization, Passive/veterinary
7.
Breast Cancer Res Treat ; 180(3): 623, 2020 04.
Article in English | MEDLINE | ID: mdl-32180072

ABSTRACT

In the original publication of the article, the spelling of the sixth author's given name was incorrect. The corrected author name should read as "Wadie David". The original article has been corrected.

8.
Breast Cancer Res Treat ; 180(3): 611-622, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32067153

ABSTRACT

PURPOSE: Studies have identified several estrogen receptor α (ERα) ligand-binding domain (LBD) somatic mutations in endocrine therapy resistant, metastatic ER-positive breast cancers. The most common mutations, Tyr537Ser (Y537S) and Asp538Gly (D538G), are detected in ~ 30% of endocrine resistant metastatic breast cancer patients. These ESR1 mutations induce the agonist conformation of ERα, confer an estrogen-independent phenotype, and promote drug resistance to antiestrogens. METHODS: ER-positive, estrogen-dependent MCF-7 cells were engineered to express either the Y537S or D538G mutants using CRISPR knock-in (cY537S and cD538G). These cells were used to screen several estrogen receptor degrader (ERD) compounds synthesized using the Proteolysis Targeting Chimeras (PROTAC) method to induce degradation of ERα via the ubiquitin-proteasome pathway. RESULTS: Wild-type MCF-7 and ERα LBD mutant cells were treated with ERD-148 (10 pM-1 µM) and assayed for cellular proliferation using the PrestoBlue cell viability assay. ERD-148 attenuated ER-dependent growth with IC50 values of 0.8, 10.5, and 6.1 nM in MCF-7, cY537S, and cD538G cells, respectively. Western blot analysis showed that MCF-7 cells treated with 1 nM ERD-148 for 24 h exhibited reduced ERα protein expression as compared to the mutants. The ER-regulated gene, GREB1, demonstrated significant downregulation in parental and mutant cells after 24 h of ERD-148 treatment at 10 nM. Growth of the ER-negative, estrogen-independent MDA-MB-231 breast cancer cells was not inhibited by ERD-148 at the ~ IC90 observed in the ER-positive cells. CONCLUSION: ERD-148 inhibits the growth of ER-positive breast cancer cells via downregulating ERα with comparable potency to Fulvestrant with marginal non-specific toxicity.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Mutation , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Proliferation/drug effects , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Humans , Proteolysis , Tumor Cells, Cultured
9.
Toxicology ; 421: 41-48, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30940549

ABSTRACT

Breast cancer is the most diagnosed malignancy among women in the United States. Approximately 70% of breast tumors express estrogen receptor alpha and are deemed ER-positive. ER-positive breast tumors depend upon endogenous estrogens to promote ER-mediated cellular proliferation. Decades of research have led to a fundamental understanding of the role ER signaling in this disease and this knowledge has led to significant advancements in the clinical use of antiestrogens for breast cancer treatment. However, adjuvant breast cancer recurrence and metastatic disease progression due to endocrine therapy resistance are prominent and unresolved issues. The established role that estrogens play in breast cancer pathogenesis explains why some patients initially respond to endocrine therapy but also why a significant number of patients become refractory to antiestrogen treatment. It is been hypothesized that exposure to environmental steroid hormone mimics and/or acquired mechanisms of resistance may explain why endocrine therapy fails in a subset of breast cancer patients. This review will highlight: 1) the relationship between ER signaling and breast cancer pathogenesis, 2) the implication of environmental exposures on steroid hormone regulated processes including breast cancer, and 3) the unresolved issue of endocrine therapy resistance.


Subject(s)
Breast Neoplasms , Endocrine Disruptors , Environmental Pollutants , Estrogens , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Disease Progression , Environmental Exposure , Estrogens/metabolism , Humans , Receptors, Estrogen/metabolism , Risk Factors , Signal Transduction
10.
Comput Toxicol ; 10: 1-16, 2019 May.
Article in English | MEDLINE | ID: mdl-30740556

ABSTRACT

Crystal structures exist for human, but not rodent, estrogen receptor-α ligand-binding domain (ERα-LBD). Consequently, rodent studies involving binding of compounds to ERα-LBD are limited in their molecular-level interpretation and extrapolation to humans. Because the sequences of rodent and human ERα-LBDs are > 95% identical, we expected their 3D structures and ligand binding to be highly similar. To test this hypothesis, we used the human ERα-LBD structure (PDB 3UUD) as a template to produce rat and mouse homology models. Employing the rodent models and human structure, we generated docking poses of 23 Group A ligands (17ß-estradiol, diethylstilbestrol, and 21 paraben analogs) in AutoDock Vina for interspecies comparisons. Ligand RMSDs (Å) (median, 95% CI) were 0.49 (0.21-1.82) (human-mouse) and 1.19 (0.22-1.82) (human-rat), well below the 2.0-2.5 Å range for equivalent docking poses. Numbers of interspecies ligand-receptor residue contacts were highly similar, with Sorensen Sc (%) = 96.8 (90.0-100) (human-mouse) and 97.7 (89.5-100) (human-rat). Likewise, numbers of interspecies ligand-receptor residue contacts were highly correlated: Pearson r = 0.913 (human-mouse) and 0.925 (human-rat). Numbers of interspecies ligand-receptor atom contacts were even more tightly correlated: r = 0.979 (human-mouse) and 0.986 (human-rat). Pyramid plots of numbers of ligand-receptor atom contacts by residue exhibited high interspecies symmetry and had Spearman r s = 0.977 (human-mouse) and 0.966 (human-rat). Group B ligands included 15 ring-substituted parabens recently shown experimentally to exhibit decreased binding to human ERα and to exert increased antimicrobial activity. Ligand efficiencies calculated from docking ligands into human ERα-LBD were well correlated with those derived from published experimental data (Pearson partial r p = 0.894 and 0.918; Groups A and B, respectively). Overall, the results indicate that our constructed rodent ERα-LBDs interact with ligands in like manner to the human receptor, thus providing a high level of confidence in extrapolations of rodent to human ligand-receptor interactions.

11.
J Women Aging ; 31(6): 553-565, 2019.
Article in English | MEDLINE | ID: mdl-30295566

ABSTRACT

The clinical efficacy of anti-osteoporotic treatments in old patients is discussed. The aim of this study was to assess if the use of anti-osteoporotic treatments for the secondary prevention of osteoporotic fractures could reduce the risk of refractures in patients over 75 years old in a Fracture Liaison Service. In this population of frail, elderly patients presenting with a recent osteoporotic fracture, we observed that the refracture incidence was similar in the treated group and the untreated group during the first year. However, 30 months after the index fracture, the osteoporosis medication for a year or more reduced the incidence of refractures by 70%.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Osteoporosis/drug therapy , Osteoporotic Fractures/prevention & control , Secondary Prevention/methods , Aged , Aged, 80 and over , Female , Frail Elderly , Humans , Incidence , Male , Osteoporosis/complications , Osteoporotic Fractures/epidemiology , Prospective Studies , Time Factors , Treatment Outcome
12.
Toxicol Sci ; 164(1): 50-59, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29945225

ABSTRACT

Two oxidized metabolites of n-butylparaben (BuP) and iso-butylparaben (IsoBuP) discovered in human urine samples exhibit structural similarity to endogenous estrogens. We hypothesized that these metabolites bind to the human estrogen receptor (ER) and promote estrogen signaling. We tested this using models of ER-mediated cellular proliferation. The estrogenic properties of 3-hydroxy n-butyl 4-hydroxybenzoate (3OH) and 2-hydroxy iso-butyl 4-hydroxybenzoate (2OH) were determined using the ER-positive, estrogen-dependent human breast cancer cell lines MCF-7, and T47D. The 3OH metabolite induced cellular proliferation with EC50 of 8.2 µM in MCF-7 cells. The EC50 for 3OH in T47D cells could not be reached. The 2OH metabolite induced proliferation with EC50 of 2.2 µM and 43.0 µM in MCF-7 and T47D cells, respectively. The EC50 for the parental IsoBuP and BuP was 0.30 and 1.2 µM in MCF-7 cells, respectively. The expression of a pro-proliferative, estrogen-inducible gene (GREB1) was induced by these compounds and blocked by co-administration of an ER antagonist (ICI 182, 780), confirming the ER-dependence of these effects. The metabolites promoted significant ER-dependent transcriptional activity of an ERE-luciferase reporter construct at 10 and 20 µM for 2OH and 10 µM for 3OH. Computational docking studies showed that the paraben compounds exhibited the potential for favorable ligand-binding domain interactions with human ERα in a manner similar to known x-ray crystal structures of 17ß-estradiol in complex with ERα. We conclude that the hydroxylated metabolites of BuP and IsoBuP are weak estrogens and should be considered as additional components of potential endocrine disrupting effects upon paraben exposure.


Subject(s)
Cell Proliferation/drug effects , Endocrine Disruptors/toxicity , Estrogen Receptor alpha/metabolism , Parabens/toxicity , Cell Survival/drug effects , Endocrine Disruptors/metabolism , Gene Expression/drug effects , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Neoplasm Proteins/genetics , Parabens/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...