Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Int Med Res ; 51(9): 3000605231190553, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37756600

ABSTRACT

We report the clinical features and genetic testing of a child with Smith-Magenis syndrome (SMS) to improve the understanding of this disease. The clinical data and molecular genetic test results of a child with SMS caused by a novel mutation in the retinoic acid-induced-1 (RAI1) gene were reviewed. A female patient aged 12 years and 9 months presented to the clinic because her mental and motor development was lagging behind that of her peers. The child had learning difficulties, poor motor coordination, temper tantrums, and self-injurious behaviors, such as skin scratching. She had a peculiar facial appearance, dry skin with scattered eczema, low hairline, wide forehead, flat face, collapsed nasal bridge, turned out upper lip, and deep palmar lines on the right hand through the palm. Wechsler's IQ test score was 48. Her electroencephalogram was normal. The diagnosis of SMS was confirmed by a heterozygous mutation in exon 3 of the RAI1 gene on chromosome chr-1717696650 at locus c.388C>T (P.Q130X). In addition, this patient had severe eczema on the skin. The RAI1 mutation c.388C>T (P.Q130X) is a newly reported variant that will help in the clinical identification of SMS and the precise localization of more phenotypically related genes.


Subject(s)
Eczema , Smith-Magenis Syndrome , Humans , Child , Female , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Transcription Factors/genetics , Trans-Activators/genetics , Phenotype , Mutation/genetics
2.
Cell Death Dis ; 13(11): 981, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36411275

ABSTRACT

Smith-Magenis syndrome (SMS) is a neurodevelopmental disorder characterized by cognitive and behavioral symptoms, obesity, and sleep disturbance, and no therapy has been developed to alleviate its symptoms or delay disease onset. SMS occurs due to haploinsufficiency of the retinoic acid-induced-1 (RAI1) gene caused by either chromosomal deletion (SMS-del) or RAI1 missense/nonsense mutation. The molecular mechanisms underlying SMS are unknown. Here, we generated and characterized primary cells derived from four SMS patients (two with SMS-del and two carrying RAI1 point mutations) and four control subjects to investigate the pathogenetic processes underlying SMS. By combining transcriptomic and lipidomic analyses, we found altered expression of lipid and lysosomal genes, deregulation of lipid metabolism, accumulation of lipid droplets, and blocked autophagic flux. We also found that SMS cells exhibited increased cell death associated with the mitochondrial pathology and the production of reactive oxygen species. Treatment with N-acetylcysteine reduced cell death and lipid accumulation, which suggests a causative link between metabolic dyshomeostasis and cell viability. Our results highlight the pathological processes in human SMS cells involving lipid metabolism, autophagy defects and mitochondrial dysfunction and suggest new potential therapeutic targets for patient treatment.


Subject(s)
Smith-Magenis Syndrome , Humans , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/pathology , Haploinsufficiency/genetics , Lipid Metabolism/genetics , Transcription Factors/metabolism , Trans-Activators/metabolism , Phenotype , Autophagy/genetics , Tretinoin/pharmacology , Tretinoin/metabolism , Lipids
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(9): 1005-1010, 2022 Sep 10.
Article in Chinese | MEDLINE | ID: mdl-36082575

ABSTRACT

OBJECTIVE: To report on a case of Smith-Magenis syndrome (SMS) due to a rare small-scale deletion. METHODS: Muscle samples from the the third fetus was collected after the in Medical history and clinical data of the patient were collected. The child and his parents were subjected to chromosome karyotyping analysis, multiplex ligation-dependent probe amplification (MLPA) and copy number variation sequencing (CNV-seq). RESULTS: The child was found to have a normal karyotype. MLPA and CNV-seq detection showed that he has harbored a 1.22 Mb deletion and a 0.3 Mb duplication in the 17p11.2 region. Neither of his parents was found to have similar deletion or duplication. CONCLUSION: The child was diagnosed with SMS due to a rare 1.22 Mb deletion in the 17p11.2 region, which is among the smallest deletions associated with this syndrome.


Subject(s)
Abnormalities, Multiple , Intellectual Disability , Smith-Magenis Syndrome , Abnormalities, Multiple/genetics , Child , Chromosome Deletion , Chromosomes, Human, Pair 17 , DNA Copy Number Variations , Humans , Intellectual Disability/genetics , Male , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics
4.
Eur J Hum Genet ; 30(11): 1233-1238, 2022 11.
Article in English | MEDLINE | ID: mdl-35821519

ABSTRACT

RAI1 is a dosage-sensitive gene whose decreased or increased expression by recurrent and non-recurrent 17p11.2 deletions or duplications causes Smith-Magenis (SMS) or Potocki-Lupski syndromes (PTLS), respectively. Here we report on a 21-year-old female patient showing SMS phenotype who was found to carry a 3.4 kb de novo intragenic RAI1 deletion. Interestingly, a significant increase in RAI1 transcript levels was identified in the patient's, brother's and mother's peripheral blood cells. Allele-specific dosage analysis revealed that the patient's maternally inherited overexpressed RAI1 allele harbors the intragenic deletion, confirming the SMS diagnosis due to the presence of a single wild-type RAI1 functional allele. The mother and brother do not present any PTLS neurologic/behavioral clinical features. Extensive sequencing of RAI1 promoter and predicted regulatory regions showed no potential causative variants accounting for gene overexpression. However, the mother and both children share a novel private missense variant in RAI1 exon 3, currently classified as a VUS (uncertain significance), though predicted by two bioinformatic tools to disrupt the binding site of one specific transcription factor. The reported familial case, the second showing RAI1 overexpression in the absence of RAI1 duplication, may help to understand the regulation of RAI1 dosage sensitivity although its phenotypic effect remains to be determined.


Subject(s)
Abnormalities, Multiple , Smith-Magenis Syndrome , Male , Female , Humans , Trans-Activators/genetics , Alleles , Maternal Inheritance , Abnormalities, Multiple/genetics , Phenotype , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics
5.
Am J Med Genet A ; 188(7): 2184-2186, 2022 07.
Article in English | MEDLINE | ID: mdl-35373511

ABSTRACT

Smith Magenis syndrome (SMS) is a rare neurobehavioral disorder caused by 17p11.2 microdeletion encompassing Retinoic Acid-Induced 1 (RAI1) gene (90% of cases) or by RAI1 point mutation (10% of cases). The neuropsychological phenotype of individuals with 17p11.2 deletion and in those with RAI1 variants mostly overlaps. However, cardiac defects have been described only in patients with a deletion so far. Here, we present the first case of a patient affected by SMS caused by RAI1 variant in whom a severe congenital pulmonary valve stenosis was diagnosed at birth, requiring trans catheter dilatation in the first month of life. This case expands the phenotypic spectrum associated with RAI1 variants in SMS, describing a previously unreported association with a congenital heart disease.


Subject(s)
Heart Defects, Congenital , Smith-Magenis Syndrome , Chromosomes, Human, Pair 17 , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Humans , Mutation , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Trans-Activators/genetics , Transcription Factors/genetics
7.
Genes (Basel) ; 13(2)2022 02 11.
Article in English | MEDLINE | ID: mdl-35205380

ABSTRACT

Smith-Magenis syndrome (SMS) is a complex genetic disorder characterized by distinctive physical features, developmental delay, cognitive impairment, and a typical behavioral phenotype. SMS is caused by interstitial 17p11.2 deletions (90%), encompassing multiple genes and including the retinoic acid-induced 1 gene (RAI1), or by pathogenic variants in RAI1 itself (10%). RAI1 is a dosage-sensitive gene expressed in many tissues and acting as transcriptional regulator. The majority of individuals exhibit a mild-to-moderate range of intellectual disability. The behavioral phenotype includes significant sleep disturbance, stereotypes, maladaptive and self-injurious behaviors. In this review, we summarize current clinical knowledge and therapeutic approaches. We further discuss the common biological background shared with other conditions commonly retained in differential diagnosis.


Subject(s)
Intellectual Disability , Smith-Magenis Syndrome , Humans , Intellectual Disability/genetics , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Trans-Activators , Transcription Factors/genetics
9.
Clin Genet ; 99(4): 519-528, 2021 04.
Article in English | MEDLINE | ID: mdl-33368193

ABSTRACT

Smith-Magenis syndrome (SMS), characterized by dysmorphic features, neurodevelopmental disorder, and sleep disturbance, is due to an interstitial deletion of chromosome 17p11.2 (90%) or to point mutations in the RAI1 gene. In this retrospective cohort, we studied the clinical, cognitive, and behavioral profile of 47 European patients with SMS caused by a 17p11.2 deletion. We update the clinical and neurobehavioral profile of SMS. Intrauterine growth was normal in most patients. Prenatal anomalies were reported in 15%. 60% of our patients older than 10 years were overweight. Prevalence of heart defects (6.5% tetralogy of Fallot, 6.5% pulmonary stenosis), ophthalmological problems (89%), scoliosis (43%), or deafness (32%) were consistent with previous reports. Epilepsy was uncommon (2%). We identified a high prevalence of obstipation (45%). All patients had learning difficulties and developmental delay, but ID range was wide and 10% of patients had IQ in the normal range. Behavioral problems included temper tantrums and other difficult behaviors (84%) and night-time awakenings (86%). Optimal care of SMS children is multidisciplinary and requires important parental involvement. In our series, half of patients were able to follow adapted schooling, but 70% of parents had to adapt their working time, illustrating the medical, social, educative, and familial impact of having a child with SMS.


Subject(s)
Smith-Magenis Syndrome/epidemiology , Abnormalities, Multiple/genetics , Adolescent , Child , Child Behavior Disorders/genetics , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 17/ultrastructure , Developmental Disabilities/genetics , Education, Special , Family Relations , Growth Disorders/genetics , Humans , Intellectual Disability/genetics , Overweight/genetics , Parents , Patient Acceptance of Health Care/statistics & numerical data , Phenotype , Prenatal Diagnosis , Retrospective Studies , Sleep Wake Disorders/genetics , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/embryology , Smith-Magenis Syndrome/psychology , Young Adult
10.
Dtsch Med Wochenschr ; 145(16): 1139-1144, 2020 08.
Article in German | MEDLINE | ID: mdl-32791549

ABSTRACT

Two major advances were made in the treatment of chronic lymphocytic leukaemia (CLL): the addition of the antibody rituximab to chemotherapy two decades ago and the introduction of the targeted agents during the last few years. Four targeted drugs with different mechanisms of action were added to the armamentarium of CLL treatment: the anti-CD20 antibody obinutuzumab, the two kinase inhibitors ibrutinib and idelalisib, which target the Bruton tyrosine kinase (BTK) and Phosphatidylinositiol-3-Kinase (PI3K) respectively in the B-cell receptor signalling pathway, as well as the Bcl2-antagonist venetoclax.Recently, the combination of venetoclax/obinutuzumab was approved for the first-line treatment of all CLL patients based on a phase-III trial in elderly unfit patients. This combination was shown to be clearly superior to chlorambucil/obinutuzumab and should become the preferred first-line treatment for the so called "slow-go" patients. Other options for these elderly, unfit patients are continuous ibrutinib or chlorambucil/obinutuzumab. Although data from phase-III studies are not yet available, venetoclax/obintuzumab may also be offered to younger, fit patients. Established therapeutic options for these so called "go go" patients are ibrutinib, fludarabin/cyclophosphamide/rituximab or bendemustine/rituximab (if > 65 years). Patients with the high-risk parameters deletion 17p or TP53mutation are known to poorly respond to chemo(immuno)therapy and should receive either ibrutinib or venetoclax/obinutuzumab.Thus, a choice has to be made between a continuous monotherapy with ibrutinib or a time-limited combination with either venetoclax/obinutuzumab (12 months) or chemoimmunotherapy (usually 6 months). In addition to disease-related factors (e. g. presence of deletion 17p/TP53 mutation, IgHV mutational status, prior therapies), comorbidities, co-medication and the specific side effects of the CLL therapies (myelosuppression, infections and secondary malignancies for chemoimmunotherapy; cardiac toxicity, bleeding and autoimmune disease for ibrutinib; tumour-lysis syndromes and infections for venetoclax) the patient's expectations need to be considered.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Adenine/adverse effects , Adenine/analogs & derivatives , Adenine/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Comorbidity , Drug Approval , Humans , Immunotherapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Staging , Piperidines/adverse effects , Piperidines/therapeutic use , Prognosis , Purines/adverse effects , Purines/therapeutic use , Quinazolinones/adverse effects , Quinazolinones/therapeutic use , Rituximab/adverse effects , Rituximab/therapeutic use , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/drug therapy , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/pathology , Sulfonamides/adverse effects , Sulfonamides/therapeutic use
11.
J Genet ; 992020.
Article in English | MEDLINE | ID: mdl-32661203

ABSTRACT

Smith-Magenis syndrome (SMS, OMIM: 182290) is a multiple congenital anomalies and intellectual disability syndrome due to a 3.45 Mb microdeletion involving 17p11.2 and is estimated to occur about one in 25,000 births. Up to now, the ultrasound findings of the foetus with SMS and their external genital defects in patients are rarely reported. This case indicates that foetus with SMS may presentpolyhydramnios and ventriculomegaly in the second trimester. The newborn male patient had an abnormal phenotype in which he has micropenis and his anus is close to the perineal body. The identification of this case may further expand the phenotypic spectrum of this genetic disorder.


Subject(s)
Abnormalities, Multiple/diagnosis , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Genitalia/abnormalities , Prenatal Diagnosis/methods , Smith-Magenis Syndrome/diagnosis , Abnormalities, Multiple/genetics , Female , Humans , Infant, Newborn , Male , Phenotype , Pregnancy , Smith-Magenis Syndrome/genetics
12.
Ann Hematol ; 99(7): 1551-1560, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32504186

ABSTRACT

TP53 aberrations reportedly predict favorable responses to decitabine (DAC) in acute myeloid leukemia (AML). We evaluated clinical features and outcomes associated with chromosome 17p loss or TP53 gene mutations in older, unfit DAC-treated AML patients in a phase II trial. Of 178 patients, 25 had loss of 17p in metaphase cytogenetics; 24 of these had a complex (CK+) and 21 a monosomal karyotype (MK+). In analyses in all patients and restricted to CK+ and MK+ patients, 17p loss tended to associate with higher rates of complete remission (CR), partial remission (PR), or antileukemic effect (ALE). Despite favorable response rates, there was no significant OS difference between patients with or without loss of 17p in the entire cohort or in the CK+ and MK+ cohort. TP53 mutations were identified in eight of 45 patients with material available. Five of the eight TP53-mutated patients had 17p loss. TP53-mutated patients had similar rates of CR/PR/ALE but shorter OS than those with TP53 wild type (P = 0.036). Moreover, patients with a subclone based on mutation data had shorter OS than those without (P = 0.05); only one patient with TP53-mutated AML had a subclone. In conclusion, 17p loss conferred a favorable impact on response rates, even among CK+ and MK+ patients that however could not be maintained. The effect of TP53 mutations appeared to be different; however, patient numbers were low. Future research needs to further dissect the impact of the various TP53 aberrations in HMA-based combination therapies. The limited duration of favorable responses to HMA treatment in adverse-risk genetics AML should prompt physicians to advance allografting for eligible patients in a timely fashion.


Subject(s)
Chromosome Deletion , Decitabine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Monosomy , Smith-Magenis Syndrome , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Chromosomes, Human, Pair 17/genetics , Clonal Evolution/drug effects , Clonal Evolution/genetics , DNA Mutational Analysis , Female , Germany/epidemiology , Humans , Karyotype , Karyotyping , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Monosomy/diagnosis , Monosomy/genetics , Mutation , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/epidemiology , Smith-Magenis Syndrome/genetics , Survival Analysis
13.
Am J Med Genet A ; 182(7): 1791-1795, 2020 07.
Article in English | MEDLINE | ID: mdl-32359026

ABSTRACT

A rare developmental delay (DD)/intellectual disability (ID) syndrome with craniofacial dysmorphisms and autistic features, termed White-Sutton syndrome (WHSUS, MIM#614787), has been recently described, identifying truncating mutations in the chromatin regulator POGZ (KIAA0461, MIM#614787). We describe a further WHSUS patient harboring a novel nonsense de novo POGZ variant, which afflicts a protein domain with transposase activity less frequently impacted by mutational events (DDE domain). This patient displays additional physical and behavioral features, these latter mimicking Smith-Magenis syndrome (SMS, MIM#182290). Considering sleep-wake cycle anomalies and abnormal behavior manifested by this boy, we reinforced the clinical resemblance between WHSUS and SMS, being both chromatinopathies. In addition, using the DeepGestalt technology, we identified a different facial overlap between WHSUS patients with mutations in the DDE domain (Group 1) and individuals harboring variants in other protein domains/regions (Group 2). This report further delineates the clinical and molecular repertoire of the POGZ-related phenotype, adding a novel patient with uncommon clinical and behavioral features and provides the first computer-aided facial study of WHSUS patients.


Subject(s)
Developmental Disabilities/genetics , Intellectual Disability/genetics , Smith-Magenis Syndrome/genetics , Transposases/genetics , Child, Preschool , Codon, Nonsense/genetics , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/diagnostic imaging , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/physiopathology , Developmental Disabilities/diagnosis , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/physiopathology , Exome/genetics , Female , Humans , Intellectual Disability/diagnosis , Intellectual Disability/diagnostic imaging , Intellectual Disability/physiopathology , Male , Mutation/genetics , Phenotype , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/diagnostic imaging , Smith-Magenis Syndrome/physiopathology
14.
Leuk Res ; 91: 106335, 2020 04.
Article in English | MEDLINE | ID: mdl-32114372

ABSTRACT

We performed a retrospective study comparing treatment patterns and overall survival (OS) in chronic lymphocytic leukemia (CLL) patients with the advent of ibrutinib to provide current real-world data. METHODS: Using a provincial population-based database, we analyzed CLL patients who received upfront treatment in British Columbia before ibrutinib availability (1984-2014), during ibrutinib access for: relapse only (2014-2015) and for upfront treatment of patients (with 17p deletion or unfit for chemotherapy) (2015-2016). Analysis included up to third-line treatment. RESULTS: Of 1729 patients meeting inclusion criteria (median age, 66 years; 1466, period 1; 140, period 2; 123, period 3), FR was the most common first-line therapy (35.8 %, 54.3 % and 40.7 %, periods 1-3, respectively) and 18.7 % received ibrutinib upfront in period 3. The most common therapies in relapse were chemoimmunotherapy (36.1 % and 55.6 %, periods 1 and 2, second-line; 29.2 %, period 1, third-line) and ibrutinib (69.8 %, period 3, second-line; 46.4 % and 70.3 %, periods 2 and 3, third-line). OS improved for patients treated in periods 2-3 over period 1 (median OS not reached vs. 11.9 years, p < 0.001; no difference in OS for periods 2-3, p = 0.385). CONCLUSION: Ibrutinib has replaced chemoimmunotherapy as the preferred therapy in relapse. Overall survival has improved over time with access to ibrutinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Smith-Magenis Syndrome/therapy , Adenine/analogs & derivatives , Adult , Aged , Aged, 80 and over , British Columbia , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Cyclophosphamide/therapeutic use , Disease Management , Doxorubicin/therapeutic use , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Neoplasm, Residual , Piperidines , Prednisone/therapeutic use , Prognosis , Recurrence , Remission Induction , Retrospective Studies , Rituximab/therapeutic use , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/mortality , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Vincristine/therapeutic use
15.
Sci Rep ; 10(1): 5009, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32193467

ABSTRACT

DNA copy number aberrations (CNA) are frequently observed in colorectal cancers (CRC). There is an urgent need for CNA-based biomarkers in clinics,. n For Stage III CRC, if combined with imaging or pathologic evidence, these markers promise more precise care. We conducted this Stage III specific biomarker discovery with a cohort of 134 CRCs, and with a newly developed high-efficiency CNA profiling protocol. Specifically, we developed the profiling protocol for tumor-normal matched tissue samples based on low-coverage clinical whole-genome sequencing (WGS). We demonstrated the protocol's accuracy and robustness by a systematic benchmark with microarray, high-coverage whole-exome and -genome approaches, where the low-coverage WGS-derived CNA segments were highly accordant (PCC >0.95) with those derived from microarray, and they were substantially less variable if compared to exome-derived segments. A lasso-based model and multivariate cox regression analysis identified a chromosome 17p loss, containing the TP53 tumor suppressor gene, that was significantly associated with reduced survival (P = 0.0139, HR = 1.688, 95% CI = [1.112-2.562]), which was validated by an independent cohort of 187 Stage III CRCs. In summary, this low-coverage WGS protocol has high sensitivity, high resolution and low cost and the identified 17p-loss is an effective poor prognosis marker for Stage III patients.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Copy Number Variations/genetics , Gene Deletion , Tumor Suppressor Protein p53/genetics , Whole Genome Sequencing/methods , Adult , Aged , Aged, 80 and over , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Female , Genetic Markers , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Survival Rate , Young Adult
16.
Br J Haematol ; 188(5): 652-660, 2020 03.
Article in English | MEDLINE | ID: mdl-30836431

ABSTRACT

Genomic technologies are revolutionizing the practice of haematology-oncology, leading to improved disease detection, more accurate prognostication and targeted treatment decisions. These advances, however, have also introduced new clinical challenges, which include problems of prognostic underdetermination and its attendant risks of over- and undertreatment. Genomic data is generated from different technologies, from cytogenetics to next-generation sequencing, which are often interpreted interchangeably and in a binary fashion-as the presence or absence of a given chromosomal deletion or mutation-an oversimplification which may lead to mistaken prognosis. We discuss the clinical use of one such prognostic marker, represented by sequence and copy number alterations in TP53, located on chromosome 17p. Mutations in TP53 are strongly linked to poor prognosis in a variety of haematological malignancies, including chronic lymphocytic leukaemia (CLL). We review studies in CLL which utilize the 17p deletion or TP53 mutations for prognostic stratification with specific focus on the technologies used for detection, the thresholds established for clinical significance, and the clinical contexts in which these alterations are identified. The case of CLL illustrates issues arising from simplistic, binary interpretation of genetic testing and highlights the need to apply a critical lens when incorporating genomics into prognostic models.


Subject(s)
Databases, Genetic , Genomics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Models, Genetic , Smith-Magenis Syndrome/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Prognosis , Smith-Magenis Syndrome/diagnosis , Tumor Suppressor Protein p53
18.
Int J Mol Sci ; 20(14)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31330985

ABSTRACT

Smith-Magenis syndrome (SMS), linked to Retinoic Acid Induced (RAI1) haploinsufficiency, is a unique model of the inversion of circadian melatonin secretion. In this regard, this model is a formidable approach to better understand circadian melatonin secretion cycle disorders and the role of the RAI1 gene in this cycle. Sleep-wake cycle disorders in SMS include sleep maintenance disorders with a phase advance and intense sleepiness around noon. These disorders have been linked to a general disturbance of sleep-wake rhythm and coexist with inverted secretion of melatonin. The exact mechanism underlying the inversion of circadian melatonin secretion in SMS has rarely been discussed. We suggest three hypotheses that could account for the inversion of circadian melatonin secretion and discuss them. First, inversion of the circadian melatonin secretion rhythm could be linked to alterations in light signal transduction. Second, this inversion could imply global misalignment of the circadian system. Third, the inversion is not linked to a global circadian clock shift but rather to a specific impairment in the melatonin secretion pathway between the suprachiasmatic nuclei (SCN) and pinealocytes. The development of diurnal SMS animal models that produce melatonin appears to be an indispensable step to further understand the molecular basis of the circadian melatonin secretion rhythm.


Subject(s)
Disease Susceptibility , Melatonin/biosynthesis , Smith-Magenis Syndrome/etiology , Smith-Magenis Syndrome/metabolism , Animals , Chromosome Mapping , Circadian Rhythm , Genetic Predisposition to Disease , Humans , Quantitative Trait Loci , Quantitative Trait, Heritable , Smith-Magenis Syndrome/diagnosis
19.
Curr Opin Psychiatry ; 32(2): 73-78, 2019 03.
Article in English | MEDLINE | ID: mdl-30557269

ABSTRACT

PURPOSE OF REVIEW: To provide an update of the most recent studies on Smith-Magenis syndrome (SMS) with a focus on the unique pattern of behavioral and sleep disturbances associated with the condition. RECENT FINDINGS: The recent literature on SMS has focused on the characteristic severe behavioral and sleep disturbances. A better understanding of the underlying pathophysiological mechanisms and common clinical course has helped further characterize SMS, while much is left to be discovered in regard to effective treatment/management. SUMMARY: SMS is a difficult to manage genetic condition defined by pervasive and progressive behavioral and sleep disturbances with a unique pattern that can often be easily discerned from other neurodevelopmental disorders. Common behavioral features include maladaptive/self-injurious, aggressive, stereotypic, and the newly appreciated food seeking behaviors associated with SMS. In addition, there is a sleep disturbance defined by an altered circadian rhythm with frequent nighttime waking and daytime sleepiness, causing patients and families significant distress. Small studies have suggested some treatment/management approaches to the behavioral and sleep disturbances, however, much remains to be discovered.


Subject(s)
Sleep Disorders, Circadian Rhythm , Smith-Magenis Syndrome , Aggression , Disease Progression , Female , Humans , Male , Psychopathology , Sleep Disorders, Circadian Rhythm/etiology , Sleep Disorders, Circadian Rhythm/psychology , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/physiopathology , Smith-Magenis Syndrome/psychology , Somnambulism , Stereotyped Behavior
20.
Ann Hematol ; 97(12): 2269-2278, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30315344

ABSTRACT

Chronic lymphocytic leukemia (CLL), a disorder for which B cell heterogeneity and increased cellular proliferation play central pathogenic roles, displays several genetic abnormalities that are associated with poor prognosis and have therapeutic implications. In this review, we discuss the prognostic role and therapeutic implications of chromosome 17p deletions and TP53 mutations in CLL. Unlike other recurrent genetic abnormalities, the frequency of TP53 alterations is relatively low in newly diagnosed patients, but increases sharply with disease progression, which suggests that these alterations represent an evolutionary mechanism of resistance. In comparison with patients without such abnormalities, those with 17p deletions and TP53 mutations have lower response rates and more aggressive disease. One important consequence of the diverse molecular mechanisms that affect the TP53 pathway is the need to assess both the presence of 17p deletion and TP53 mutations before treatment initiation. Several authors have attempted to incorporate TP53 abnormalities in different prognostic models for CLL, and the recent International Prognostic Index for Chronic Lymphocytic Leukemia formally considers patients with TP53 abnormalities (deletion 17p or TP53 mutation or both) as high-risk. Several novel agents may improve results in patients with CLL, including in those with TP53 mutations. Ibrutinib, idelalisib, and venetoclax have been approved in various settings and countries for treatment of CLL. Further progress in targeted therapy and judicious use of chemotherapy, monoclonal antibodies, and reduced-intensity allogeneic transplantation will provide patients with CLL in general, and those with TP53 abnormalities in particular, with a better prognosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosome Deletion , Leukemia, Lymphocytic, Chronic, B-Cell , Smith-Magenis Syndrome , Stem Cell Transplantation , Tumor Suppressor Protein p53/genetics , Allografts , Chromosomes, Human, Pair 17/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Prognosis , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...