Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Med Genet A ; 194(7): e63554, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38317562

RESUMEN

Patients with Fanconi anemia (FA) are often perceived to have poor growth when general population growth curves are utilized. We hypothesize that FA patients have unique growth and aimed to create FA-specific growth charts. Height and weight data from ages 0 to 20 years were extracted from medical records of patients treated at the Fanconi Anemia Comprehensive Care Clinic at the University of Minnesota. Height, weight, and BMI growth curves were generated and fitted to reference percentiles using the Lambda-Mu-Sigma method. FA-specific percentiles were compared to WHO standards for ages 0-2 and CDC references for ages 2-20. In FA males, the 50th height- and weight-for-age percentiles overlap with the 3rd reference percentile. In FA females, only the 50th height-for-age percentile overlaps with the 3rd reference percentile. For weight, FA females show progressive growth failure between 6 and 24 months followed by stabilization around the 50th percentile. The FA BMI-for-age percentiles show similar patterns to the weight-for-age percentiles but have different timing of onset of adiposity rebound and broader variability in females. Growth in FA patients follows a different trajectory than available normative curves. FA-specific growth charts may be useful to better guide accurate growth expectations, evaluations, and treatment.


Asunto(s)
Estatura , Índice de Masa Corporal , Peso Corporal , Anemia de Fanconi , Gráficos de Crecimiento , Humanos , Femenino , Masculino , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/patología , Anemia de Fanconi/genética , Anemia de Fanconi/fisiopatología , Niño , Adolescente , Preescolar , Lactante , Adulto Joven , Recién Nacido
2.
Orphanet J Rare Dis ; 15(1): 170, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32605631

RESUMEN

BACKGROUND: Fanconi anemia is a rare disease clinically characterized by malformations, bone marrow failure and an increased risk of solid tumors and hematologic malignancies. The only therapies available are hematopoietic stem cell transplantation for bone marrow failure or leukemia, and surgical resection for solid tumors. Therefore, there is still an urgent need for new therapeutic options. With this aim, we developed a novel high-content cell-based screening assay to identify drugs with therapeutic potential in FA. RESULTS: A TALEN-mediated FANCA-deficient U2OS cell line was stably transfected with YFP-FANCD2 fusion protein. These cells were unable to form fluorescent foci or to monoubiquitinate endogenous or exogenous FANCD2 upon DNA damage and were more sensitive to mitomycin C when compared to the parental wild type counterpart. FANCA correction by retroviral infection restored the cell line's ability to form FANCD2 foci and ubiquitinate FANCD2. The feasibility of this cell-based system was interrogated in a high content screening of 3802 compounds, including a Prestwick library of 1200 FDA-approved drugs. The potential hits identified were then individually tested for their ability to rescue FANCD2 foci and monoubiquitination, and chromosomal stability in the absence of FANCA. CONCLUSIONS: While, unfortunately, none of the compounds tested were able to restore cellular FANCA-deficiency, our study shows the potential capacity to screen large compound libraries in the context of Fanconi anemia therapeutics in an optimized and cost-effective platform.


Asunto(s)
Anemia de Fanconi , Daño del ADN , Evaluación Preclínica de Medicamentos , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Humanos
3.
J Med Genet ; 57(6): 389-399, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32015000

RESUMEN

BACKGROUND: Primary hereditary microcephaly (MCPH) comprises a large group of autosomal recessive disorders mainly affecting cortical development and resulting in a congenital impairment of brain growth. Despite the identification of >25 causal genes so far, it remains a challenge to distinguish between different MCPH forms at the clinical level. METHODS: 7 patients with newly identified mutations in CDK5RAP2 (MCPH3) were investigated by performing prospective, extensive and systematic clinical, MRI, psychomotor, neurosensory and cognitive examinations under similar conditions. RESULTS: All patients displayed neurosensory defects in addition to microcephaly. Small cochlea with incomplete partition type II was found in all cases and was associated with progressive deafness in 4 of them. Furthermore, the CDK5RAP2 protein was specifically identified in the developing cochlea from human fetal tissues. Microphthalmia was also present in all patients along with retinal pigmentation changes and lipofuscin deposits. Finally, hypothalamic anomalies consisting of interhypothalamic adhesions, a congenital midline defect usually associated with holoprosencephaly, was detected in 5 cases. CONCLUSION: This is the first report indicating that CDK5RAP2 not only governs brain size but also plays a role in ocular and cochlear development and is necessary for hypothalamic nuclear separation at the midline. Our data indicate that CDK5RAP2 should be considered as a potential gene associated with deafness and forme fruste of holoprosencephaly. These children should be given neurosensory follow-up to prevent additional comorbidities and allow them reaching their full educational potential. TRIAL REGISTRATION NUMBER: NCT01565005.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cocleares/genética , Microcefalia/genética , Proteínas del Tejido Nervioso/genética , Niño , Preescolar , Cóclea/diagnóstico por imagen , Cóclea/metabolismo , Cóclea/patología , Enfermedades Cocleares/diagnóstico por imagen , Enfermedades Cocleares/patología , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Femenino , Humanos , Hipotálamo/diagnóstico por imagen , Hipotálamo/patología , Lactante , Imagen por Resonancia Magnética , Masculino , Microcefalia/diagnóstico por imagen , Microcefalia/patología , Mutación , Neurogénesis/genética , Linaje , Retina/diagnóstico por imagen , Retina/patología
4.
J Clin Invest ; 130(3): 1377-1391, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31877112

RESUMEN

Hematopoietic stem cell (HSC) attrition is considered the key event underlying progressive BM failure (BMF) in Fanconi anemia (FA), the most frequent inherited BMF disorder in humans. However, despite major advances, how the cellular, biochemical, and molecular alterations reported in FA lead to HSC exhaustion remains poorly understood. Here, we demonstrated in human and mouse cells that loss-of-function of FANCA or FANCC, products of 2 genes affecting more than 80% of FA patients worldwide, is associated with constitutive expression of the transcription factor microphthalmia (MiTF) through the cooperative, unscheduled activation of several stress-signaling pathways, including the SMAD2/3, p38 MAPK, NF-κB, and AKT cascades. We validated the unrestrained Mitf expression downstream of p38 in Fanca-/- mice, which display hallmarks of hematopoietic stress, including loss of HSC quiescence, DNA damage accumulation in HSCs, and reduced HSC repopulation capacity. Importantly, we demonstrated that shRNA-mediated downregulation of Mitf expression or inhibition of p38 signaling rescued HSC quiescence and prevented DNA damage accumulation. Our data support the hypothesis that HSC attrition in FA is the consequence of defects in the DNA-damage response combined with chronic activation of otherwise transiently activated signaling pathways, which jointly prevent the recovery of HSC quiescence.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/metabolismo , Daño del ADN , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Transcripción Asociado a Microftalmía/metabolismo , Animales , Ácido Ascórbico , Trastornos de Fallo de la Médula Ósea/genética , Trastornos de Fallo de la Médula Ósea/patología , Línea Celular , Colecalciferol , Deshidroepiandrosterona/análogos & derivados , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/patología , Ratones , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/genética , Ácidos Nicotínicos , Extractos Vegetales , Proteínas Smad/genética , Proteínas Smad/metabolismo
5.
Toxins (Basel) ; 8(7)2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27399778

RESUMEN

Epidemiological studies have found a positive association between coffee consumption and a lower risk of cardiovascular disorders, some cancers, diabetes, Parkinson and Alzheimer disease. Coffee consumption, however, has also been linked to an increased risk of developing some types of cancer, including bladder cancer in adults and leukemia in children of mothers who drink coffee during pregnancy. Since cancer is driven by the accumulation of DNA alterations, the ability of the coffee constituent caffeic acid to induce DNA damage in cells may play a role in the carcinogenic potential of this beverage. This carcinogenic potential may be exacerbated in cells with DNA repair defects. People with the genetic disease Fanconi Anemia have DNA repair deficiencies and are predisposed to several cancers, particularly acute myeloid leukemia. Defects in the DNA repair protein Fanconi Anemia D2 (FANCD2) also play an important role in the development of a variety of cancers (e.g., bladder cancer) in people without this genetic disease. This communication shows that cells deficient in FANCD2 are hypersensitive to the cytotoxicity (clonogenic assay) and DNA damage (γ-H2AX and 53BP1 focus assay) induced by caffeic acid and by a commercial lyophilized coffee extract. These data suggest that people with Fanconi Anemia, or healthy people who develop sporadic mutations in FANCD2, may be hypersensitive to the carcinogenic activity of coffee.


Asunto(s)
Ácidos Cafeicos/toxicidad , Café/toxicidad , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Anemia de Fanconi/patología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Histonas/metabolismo , Humanos , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
6.
J Natl Cancer Inst ; 106(1): djt335, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24402422

RESUMEN

BACKGROUND: There is no method routinely used to predict response to anthracycline and cyclophosphamide-based chemotherapy in the clinic; therefore patients often receive treatment for breast cancer with no benefit. Loss of the Fanconi anemia/BRCA (FA/BRCA) DNA damage response (DDR) pathway occurs in approximately 25% of breast cancer patients through several mechanisms and results in sensitization to DNA-damaging agents. The aim of this study was to develop an assay to detect DDR-deficient tumors associated with loss of the FA/BRCA pathway, for the purpose of treatment selection. METHODS: DNA microarray data from 21 FA patients and 11 control subjects were analyzed to identify genetic processes associated with a deficiency in DDR. Unsupervised hierarchical clustering was then performed using 60 BRCA1/2 mutant and 47 sporadic tumor samples, and a molecular subgroup was identified that was defined by the molecular processes represented within FA patients. A 44-gene microarray-based assay (the DDR deficiency assay) was developed to prospectively identify this subgroup from formalin-fixed, paraffin-embedded samples. All statistical tests were two-sided. RESULTS: In a publicly available independent cohort of 203 patients, the assay predicted complete pathologic response vs residual disease after neoadjuvant DNA-damaging chemotherapy (5-fluorouracil, anthracycline, and cyclophosphamide) with an odds ratio of 3.96 (95% confidence interval [Cl] =1.67 to 9.41; P = .002). In a new independent cohort of 191 breast cancer patients treated with adjuvant 5-fluorouracil, epirubicin, and cyclophosphamide, a positive assay result predicted 5-year relapse-free survival with a hazard ratio of 0.37 (95% Cl = 0.15 to 0.88; P = .03) compared with the assay negative population. CONCLUSIONS: A formalin-fixed, paraffin-embedded tissue-based assay has been developed and independently validated as a predictor of response and prognosis after anthracycline/cyclophosphamide-based chemotherapy in the neoadjuvant and adjuvant settings. These findings warrant further validation in a prospective clinical study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Daño del ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Anemia de Fanconi/metabolismo , Adulto , Anciano , Antraciclinas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/genética , Quimioterapia Adyuvante , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Epirrubicina/administración & dosificación , Anemia de Fanconi/genética , Femenino , Fluorouracilo/administración & dosificación , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Oportunidad Relativa , Análisis de Secuencia por Matrices de Oligonucleótidos , Estudios Prospectivos
7.
Int J Cancer ; 124(4): 783-92, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19048618

RESUMEN

The Fanconi Anemia (FA) DNA damage response pathway is involved in the processing of DNA interstrand crosslinks (ICLs). As such, inhibition of the FA pathway could chemosensitize FA-competent tumor cells to commonly used ICL agents like cisplatin. Moreover, suppression of the FA pathway is synthetic lethal with deficiencies in several other DNA repair pathways, suggesting that FA pathway inhibitors could be used in targeted therapies against specific tumors. To identify such inhibitors, we designed a novel in vitro screening assay utilizing Xenopus egg extracts. Using the DNA-stimulated monoubiquitylation of Xenopus FANCD2 (xFANCD2-L) as readout, a chemical library screen identified DDN (2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone) as a novel and potent FA pathway inhibitor. DDN inhibited xFANCD2-L formation in a dose-dependent manner in both extracts and human cells without disruption of the upstream FA core complex. DDN also inhibited the characteristic subnuclear FANCD2 foci formation following DNA damage. Moreover, DDN displayed a greater synergistic effect with cisplatin in a FA-proficient cancer cell line compared to its FA-deficient isogenic counterpart, suggesting that DDN might be a good lead candidate as cisplatin chemosensitizer in both FA-deficient and FA-competent tumors. This system constitutes the first cell-free screening assay for identifying compounds that inhibit the FA pathway and provides a new biochemical platform for mapping the functions of its various components with specific chemical inhibitors.


Asunto(s)
Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Animales , Supervivencia Celular , Sistema Libre de Células , Cisplatino/farmacología , Reactivos de Enlaces Cruzados/farmacología , ADN/química , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Xenopus laevis
8.
Pediatr Nephrol ; 22(10): 1711-5, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17690917

RESUMEN

The Acadians were French settlers to Nova Scotia in the seventeenth century. In 1755, they were expelled by the British to various sites in the Americas, including Louisiana, where they are referred to as Cajuns. Many later migrated back to the Maritime Provinces of Canada. The objective of this study was to describe a series of pediatric patients representing an Acadian variant of Fanconi syndrome (AVFS). Nineteen children were diagnosed with AVFS between 1971 and 2006 and followed regularly. Data concerning demographics, growth, bone disease, and renal function at presentation and last observation were collected. The commonest reason for referral was assessment of genu valgum at 8.5 +/- 4.2 years (mean +/- SD) with hypophosphatemic rickets confirmed in all patients. Small-body habitus and short stature were confirmed in all patients. Therapy consisting of alkali replacement and phosphate and vitamin D supplements resulted in improvement of rickets and leg alignment but not stature (median height Z-score at presentation -2.05, range -3.6 to 0.21, vs. -2.05 at last observation, range -3.36 to 0.47). Creatinine clearance decreased (65.4 +/- 24.6 vs. 48.0 +/- 36.0 ml/min per 1.73 m(2), P < 0.05) and proteinuria increased (0.38 +/- 0.25 vs. 1.46 +/- 1.52 g/d, P < 0.05) during follow up of 8.4 +/- 6.1 years. Chronic kidney disease developed in 50% by age 13 years. No extrarenal manifestations were identified, although two patients developed lethal pulmonary fibrosis postrenal transplantation. AVFS is characterized by rickets responsive to solute therapy, short stature, and loss of renal function, with progressive proteinuria with age.


Asunto(s)
Anemia de Fanconi/genética , Variación Genética , Adolescente , Adulto , Peso al Nacer , Peso Corporal , Canadá , Niño , Preescolar , Humanos , Proteinuria/etiología , Raquitismo/epidemiología
9.
Blood ; 103(1): 128-32, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12958075

RESUMEN

Fanconi anemia (FA) is an autosomal recessive cancer susceptibility syndrome characterized by cellular sensitivity to genotoxic agents. In recent years, FA proteins have been associated with different molecules involved in signal transduction, which has raised the interest in FA-dependent signaling pathways. Here, we report that the c-Jun N-terminal kinase (JNK) fails to phosphorylate in response to UV radiation and treatment with mitomycin C in FA lymphoblast cells derived from type A patients (FA-A). Furthermore, defective kinase activity seems to be specific for JNK, because extracellular signal-regulated kinase (ERK) responded to the proper stimuli in FA-A cells. We also demonstrate that the early growth-response factor-1 (Egr-1), a JNK downstream target gene that is normally induced by genotoxic stress, is not upregulated in UV-treated FA-A cells. Moreover, FA-A cells are more sensitive to apoptosis than control lymphoblasts. Both JNK and Egr-1 may be part of a pathway triggered by FA proteins, because functional correction of FA-A cells by gene transfer restores, at least in part, JNK activation and Egr-1 expression after UV exposure. Together, our data suggest that activation of JNK and expression of Egr-1 gene in B lymphoblasts mediate a cellular response to genotoxic agents that may be induced by FA proteins.


Asunto(s)
Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteínas Inmediatas-Precoces , Factores de Transcripción/genética , Linfocitos B/metabolismo , Linfocitos B/efectos de la radiación , Secuencia de Bases , Línea Celular , ADN Complementario/genética , Regulación hacia Abajo , Proteína 1 de la Respuesta de Crecimiento Precoz , Activación Enzimática/efectos de la radiación , Proteína del Grupo de Complementación A de la Anemia de Fanconi , Prueba de Complementación Genética , Humanos , Proteínas/genética , Proteínas/metabolismo , Rayos Ultravioleta
10.
Genomics ; 67(3): 273-83, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10936049

RESUMEN

Despite the cloning of four disease-associated genes for Fanconi anemia (FA), the molecular pathogenesis of FA remains largely unknown. To study FA complementation group A using the mouse as a model system, we cloned and characterized the mouse homolog of the human FANCA cDNA. The mouse cDNA (Fanca) encodes a 161-kDa protein that shares 65% amino acid sequence identity with human FANCA. Fanca is located at the distal region of mouse chromosome 8 and has a ubiquitous pattern of expression in embryonic and adult tissues. Expression of the mouse cDNA in human FA-A cells restores the cellular drug sensitivity to normal levels. Thus, the expression pattern, protein structure, chromosomal location, and function of FANCA are conserved in the mouse. We also isolated a novel zinc finger protein, Zfp276, which has five C(2)H(2) domains. Interestingly, Zfp276 is situated in the Fanca locus, and the 3'UTR of its cDNA overlaps with the last four exons of Fanca in a tail-to-tail manner. Zfp276 is expressed in the same tissues as Fanca, but does not complement the mitomycin C (MMC)-sensitive phenotype of FA-A cells. The overlapping genomic organization between Zfp276 and Fanca may have relevance to the disease phenotype of FA.


Asunto(s)
Clonación Molecular , ADN Complementario , Proteínas de Unión al ADN , Anemia de Fanconi/genética , Proteínas/genética , Dedos de Zinc/genética , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Células Cultivadas , Cromosomas Humanos Par 8/genética , Cartilla de ADN/química , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi , Expresión Génica , Prueba de Complementación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fenotipo , Reacción en Cadena de la Polimerasa , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
12.
Carcinogenesis ; 16(3): 555-61, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7697813

RESUMEN

Fanconi anaemia (FA) is a hereditary tumour-prone disorder. FA cells exposed to DNA crosslinking agents show an increased frequency of chromosome aberrations and of deletion type mutations. The molecular basis of FA presumably is a deficiency in cellular repair of DNA adducts. In this work a shuttle vector plasmid was treated with 8-methoxypsoralen + a split dose of UVA (leading to crosslink induction), and transfected into FA lymphoblasts. The supF gene of the vector showed a mutation frequency similar to that of normal cells; however, the number of base substitutions was relatively low, whereas a high level (50%) of deletions was seen. With both normal and FA cells these deletions varied greatly in size and were randomly distributed within the supF gene. DNA cross-links were also induced using a triple helix forming 22-mer oligonucleotide linked to a psoralen molecule and being complementary to part of supF, leading to a > 30-fold increase of mutations, which were mainly position 167 single-base substitutions and showed a pattern identical to that of the normal cells. This normal response of FA cells to the site-specific DNA cross-links may reflect that not all gene sequences of FA cells are subjected to abnormal DNA repair. Alternatively, it may reflect a lower than normal genome-overall activity of a DNA cross-link repair complex, fully capable of efficiently repairing only molecules carrying relatively few adducts.


Asunto(s)
Anemia de Fanconi/genética , Vectores Genéticos , Metoxaleno/toxicidad , Secuencia de Bases , Línea Celular , ADN/efectos de los fármacos , Aductos de ADN , Análisis Mutacional de ADN , Reparación del ADN , Anemia de Fanconi/patología , Humanos , Datos de Secuencia Molecular , Terapia PUVA , Eliminación de Secuencia
13.
Hum Genet ; 60(3): 233-8, 1982.
Artículo en Inglés | MEDLINE | ID: mdl-6809593

RESUMEN

Twin and single sister chromatid exchanges (SCEs) induced by short treatments with mitomycin C (MC) and 4,5',8-trimethylpsoralen (TMP)-plus-near ultraviolet light (NUV) were analyzed in colcemid-induced endoreduplicated normal human and typical Fanconi's anemia (FA) fibroblasts with diplochromosomes. The induction rate of twin SCEs that had occurred in the first cycle (S1) after the treatment was 1.7--2.4 times higher in FA cells than in normal cells. The induction rate of single SCEs that had arisen during the second cycle (S2) long after the treatment was also much higher, though less than the twin SCE rate, in FA cells than the almost negligible rate after repair of cross-links and monoadducts in normal cells. These results in FA cells, which specifically lack the first half-excision step of the two-step cross-link repair but retain the normal monoadduct repair, indicate that MC or TMP cross-links remaining unrepaired are indeed responsible for higher inductions of twin (S1 exchange) and single SCEs (S2 exchange). Thus, these findings indicate that Shafer's model of replication bypass for cross-link-induced SCE, which predicts greatly reduced twin SCE formation in FA cells due to half cancellation, is apparently inadequate as such. We present three plausible models, incorporating the ordinary replication model, random unilateral cross-link transfer, and chromatid breakage/reunion, that can account for the probabilistic inductions of single and twin SCEs and even for no SCE formation.


Asunto(s)
Anemia Aplásica/patología , Reactivos de Enlaces Cruzados/farmacología , Intercambio Genético/efectos de los fármacos , Anemia de Fanconi/patología , Intercambio de Cromátides Hermanas/efectos de los fármacos , Células Cultivadas , Anemia de Fanconi/genética , Fibroblastos/efectos de los fármacos , Humanos , Técnicas In Vitro , Mitomicina , Mitomicinas/farmacología , Modelos Genéticos , Terapia PUVA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA