Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.420
Filtrar
2.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(3): 962-964, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38926996

RESUMEN

Studies have found that 1/3 patients with acquired aplastic anemia have shortened telomere length, and the shorter the telomere, the longer the disease course, the more prone to relapse, the lower the overall survival rate, and the higher the probability of clonal evolution. The regulation of telomere length is affected by many factors, including telomerase activity, telomerase-related genes, telomere regulatory proteins and other related factors. Telomere shortening can lead to genetic instability and increases the probability of clonal evolution in patients with acquired aplastic anemia. This article reviews the role of telomere in the clonal evolution of acquired aplastic anemia and factors affecting telomere length.


Asunto(s)
Anemia Aplásica , Homeostasis del Telómero , Anemia Aplásica/genética , Anemia Aplásica/patología , Acortamiento del Telómero , Evolución Clonal , Tasa de Supervivencia , Recurrencia , Homeostasis del Telómero/genética , Telomerasa/genética , Telomerasa/metabolismo , Inestabilidad Genómica/genética , Humanos
3.
Cell Death Dis ; 15(5): 324, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724533

RESUMEN

Severe aplastic anemia (SAA) is a rare, fatal disease characterized by severe cytopenias and loss of hematopoietic stem cells (HSCs). Immune-mediated destruction and inflammation are known drivers of SAA, however, the underlying mechanisms driving persistent inflammation are unknown. Current treatments for SAA rely on immunosuppressive therapies or HSC transplantation, however, these treatments are not always effective. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and impaired efferocytosis in SAA mice, relative to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.


Asunto(s)
Anemia Aplásica , Antígeno CD47 , Ácido Eicosapentaenoico , Animales , Anemia Aplásica/patología , Ratones , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacología , Antígeno CD47/metabolismo , Antígeno CD47/genética , Apoptosis/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Inflamación/patología , Masculino , Eferocitosis
5.
Eur J Med Genet ; 69: 104939, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38614309

RESUMEN

ADH5/ALDH2 deficiency is a rare inherited syndrome characterized by short stature, microcephaly, delayed mental development, and hematopoietic dysfunction and has recently been proposed as a disease paradigm. Acute and severe presentations include aplastic anemia, myelodysplastic syndrome, or leukemia, requiring bone marrow transplantation during childhood. Conversely, non-hematological manifestations may exhibit a prolonged and nonspecific clinical trajectory, with growth failure and developmental delay, most of which are often overlooked, particularly in patients with milder symptoms. Here, we describe the clinical course of a girl with a wide spectrum of clinical presentations, including nonspecific hematopoietic disorders, growth retardation, mild developmental delay, amblyopia, hemophagocytic lymphohistiocytosis, and verruca vulgaris, culminating in a genetic diagnosis of AMeD syndrome at 12 years of age. We also summarized the clinical manifestations of previously reported cases of AMeD syndrome. Cumulatively, 13 females and 5 males have been documented, with a cardinal triad of symptoms, aplastic anemia, short stature, and intellectual disability. Additional characteristic observations included pigmentary deposition in approximately half of the cases and skeletal difficulties in one-quarter. We propose that early diagnosis of patients who exhibit relatively mild phenotypes of skin or skeletal lesions is important for managing and improving the quality of life of patients with AMeD syndrome.


Asunto(s)
Fenotipo , Humanos , Femenino , Niño , Aldehído Deshidrogenasa Mitocondrial/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Masculino , Microcefalia/genética , Microcefalia/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Anemia Aplásica/genética , Anemia Aplásica/patología
6.
Zhonghua Yi Xue Za Zhi ; 104(7): 465-467, 2024 Feb 20.
Artículo en Chino | MEDLINE | ID: mdl-38317357

RESUMEN

Paying attention to the diagnosis and classification of acquired aplastic anemia (AA) is the basis for improving the efficacy and the guarantee for the correct exploration of the pathological mechanism, which is of great clinical and academic significance. At present, AA classification is still based on clinical characteristics, which is a historical product of academic development.It is beneficial to guide symptomatic treatment and for the onset of curative treatment. However, the clinical classification of AA cannot replace the pathological mechanism classification to guide the treatment of the root cause. The classification of the pathological mechanism of AA determines the choice of treatment strategy, and can provide a basis for the study of etiology and prevention, and is also the future research direction. Paying attention to the classification of the pathological mechanism of AA is the basis for improving the efficacy and the guarantee for the correct exploration of the pathological mechanism. Modern medicine has entered the era of "molecular targets" and "precision", and how to treat clinical classification based on clinical characteristics is an important issue faced by clinicians. When many different mechanisms of bone marrow failure isolated from AA patients can be accurately identified, that is, when the clinically diagnosed AA has been truly purified into a disease with a clear pathological mechanism, the clinical classification of AA can help to choose the root cause strategy. This article mainly focuses on how to view the clinical classification of AA for the reference of colleagues.


Asunto(s)
Anemia Aplásica , Pancitopenia , Humanos , Anemia Aplásica/diagnóstico , Anemia Aplásica/tratamiento farmacológico , Anemia Aplásica/patología
7.
Br J Haematol ; 204(5): 2086-2096, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38296352

RESUMEN

Morphological dysplasia in haematopoietic cells, defined by a 10% threshold in each lineage, is one of the diagnostic criteria for myelodysplastic neoplasms. Dysplasia limited to the erythroid lineage has also been reported in some cases of aplastic anaemia (AA); however, its significance remains unclear. We herein examined the impact of erythroid dysplasia on immunosuppressive therapy responses and survival in AA patients. The present study included 100 eligible AA patients without ring sideroblasts. Among them, 32 had dysplasia in the erythroid lineage (AA with minimal dysplasia [mini-D]). No significant sex or age differences were observed between AA groups with and without erythroid dysplasia. In severe/very severe AA and non-severe AA patients, a response to anti-thymocyte globulin + ciclosporin within 12 months was observed in 80.0% and 60.0% of AA with mini-D and 42.9% and 90.0% of those without dysplasia, with no significant difference (p = 0.29 and p = 0.24 respectively). Overall survival and leukaemia-free survival did not significantly differ between the groups. Collectively, the present results indicate that the presence of erythroid dysplasia did not significantly affect clinical characteristics or outcomes in AA patients, suggesting that its presence in AA is acceptable. Therefore, erythroid dysplasia should not exclude an AA diagnosis.


Asunto(s)
Anemia Aplásica , Sistema de Registros , Humanos , Anemia Aplásica/mortalidad , Anemia Aplásica/patología , Anemia Aplásica/tratamiento farmacológico , Femenino , Masculino , Persona de Mediana Edad , Adulto , Anciano , Adulto Joven , Células Eritroides/patología , Adolescente , Anciano de 80 o más Años
8.
Adv Sci (Weinh) ; 11(10): e2304539, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38145351

RESUMEN

Aplastic anemia (AA) is a bone marrow (BM) failure syndrome mediated by hyperactivated T-cells with heterogeneous pathogenic factors. The onset of BM failure cannot be accurately determined in humans; therefore, exact pathogenesis remains unclear. In this study, a cellular atlas and microenvironment interactions is established using unbiased single-cell RNA-seq, along with multi-omics analyses (mass cytometry, cytokine profiling, and oxidized fatty acid metabolomics). A new KIR+ CD8+ regulatory T cells (Treg) subset is identified in patients with AA that engages in immune homeostasis. Conventional CD4+ T-cells differentiate into highly differentiated T helper cells with type 2 cytokines (IL-4, IL-6, and IL-13), GM-SCF, and IL-1ß. Immunosuppressive homeostasis is impaired by enhanced apoptosis of activated Treg cells. Pathological Vδ1 cells dominated the main fraction of γδ T-cells. The B/plasma, erythroid, and myeloid lineages also exhibit substantial pathological features. Interactions between TNFSF12-TNFRSF12A, TNF-TNFRSF1A, and granzyme-gasdermin are associated with the cell death of hematopoietic stem/progenitor (HSPCs), Treg, and early erythroid cells. Ferroptosis, a major driver of HSPCs destruction, is identified in patients with AA. Furthermore, a case of twins with AA is reported to enhance the persuasiveness of the analysis. These results collectively constitute the cellular atlas and microenvironment interactions in patients with AA and provide novel insights into the development of new therapeutic opportunities.


Asunto(s)
Anemia Aplásica , Humanos , Anemia Aplásica/patología , Células de la Médula Ósea/patología , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis/fisiología , Citocinas/metabolismo
9.
Best Pract Res Clin Haematol ; 36(4): 101516, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38092475

RESUMEN

The bone marrow failure syndromes (BMFS) are a diverse group of acquired and inherited diseases which may manifest in cytopenias, haematological malignancy and/or syndromic multisystem disease. Patients with BMFS frequently experience poor outcomes, and improved treatment strategies are needed. Collation of clinical characteristics and patient outcomes in a national disease-specific registry represents a powerful tool to identify areas of need and support clinical and research collaboration. Novel treatment strategies such as gene therapy, particularly in rare diseases, will depend on the ability to identify eligible patients alongside the molecular genetic features of their disease that may be amenable to novel therapy. The Australian Aplastic Anaemia and other Bone Marrow Failure Syndromes Registry (AAR) aims to improve outcomes for all paediatric and adult patients with BMFS in Australia by describing the demographics, treatments (including supportive care) and outcomes, and serving as a resource for research and practice improvement.


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Adulto , Humanos , Niño , Anemia Aplásica/genética , Anemia Aplásica/terapia , Anemia Aplásica/patología , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/terapia , Enfermedades de la Médula Ósea/patología , Australia/epidemiología , Trastornos de Fallo de la Médula Ósea , Síndrome , Sistema de Registros
10.
Medicina (Kaunas) ; 59(12)2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38138273

RESUMEN

Bone marrow failure (BMF) syndromes are a heterogeneous group of benign hematological conditions with common clinical features including reduced bone marrow cellularity and peripheral blood cytopenias. Acquired aplastic anemia (AA) is caused by T helper(Th)1-mediated immune responses and cytotoxic CD8+ T cell-mediated autologous immune attacks against hematopoietic stem and progenitor cells (HSPCs). Interferon-γ (IFNγ), tumor necrosis factor-α, and Fas-ligand are historically linked to AA pathogenesis because they drive Th1 and cytotoxic T cell-mediated responses and can directly induce HSPC apoptosis and differentiation block. The use of omics technologies has amplified the amount of data at the single-cell level, and knowledge on AA, and new scenarios, have been opened on "old" point of view. In this review, we summarize the current state-of-art of the pathogenic role of IFNγ in AA from initial findings to novel evidence, such as the involvement of the HIF-1α pathway, and how this knowledge can be translated in clinical practice.


Asunto(s)
Anemia Aplásica , Humanos , Anemia Aplásica/etiología , Anemia Aplásica/patología , Interferón gamma , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Células de la Médula Ósea , Linfocitos T CD8-positivos/metabolismo
11.
Metabolomics ; 19(12): 94, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37975930

RESUMEN

INTRODUCTION: Idiopathic acquired aplastic anemia (AA) is a bone marrow failure disorder where aberrant T-cell functions lead to depletion of hematopoietic stem and progenitor cells in the bone marrow (BM) microenvironment. T-cells undergo metabolic rewiring, which regulates their proliferation and differentiation. Therefore, studying metabolic variation in AA patients may aid us with a better understanding of the T-cell regulatory pathways governed by metabolites and their pathological engagement in the disease. OBJECTIVE: To identify the differential metabolites in BM plasma of AA patients, AA follow-up (AAF) in comparison to normal controls (NC) and to identify potential disease biomarker(s). METHODS: The study used 1D 1H NMR Carr-Purcell-Meiboom-Gill (CPMG) spectra to identify the metabolites present in the BM plasma samples of AA (n = 40), AAF (n = 16), and NC (n = 20). Metabolic differences between the groups and predictive biomarkers were identified by using multivariate analysis and receiver operating characteristic (ROC) module of Metaboanalyst V5.0 tool, respectively. RESULTS: The AA and AAF samples were well discriminated from NC group as per Principal Component analysis (PCA). Further, we found significant alteration in the levels of 17 metabolites in AA involved in amino-acid (Leucine, serine, threonine, phenylalanine, lysine, histidine, valine, tyrosine, and proline), carbohydrate (Glucose, lactate and mannose), fatty acid (Acetate, glycerol myo-inositol and citrate), and purine metabolism (hypoxanthine) in comparison to NC. Additionally, biomarker analysis predicted Hypoxanthine and Acetate can be used as a potential biomarker. CONCLUSION: The study highlights the significant metabolic alterations in the BM plasma of AA patients which may have implication in the disease pathobiology.


Asunto(s)
Anemia Aplásica , Médula Ósea , Humanos , Médula Ósea/metabolismo , Médula Ósea/patología , Anemia Aplásica/metabolismo , Anemia Aplásica/patología , Metabolómica , Espectroscopía de Resonancia Magnética , Biomarcadores , Acetatos , Hipoxantinas
12.
Biomed Pharmacother ; 169: 115863, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37952356

RESUMEN

Acquired aplastic anemia (AA) is a bone marrow failure (BMF) disease, characterized by fatty bone marrow (BM) and BM hypocellularity resulted from auto-immune dysregulated T cells-mediated destruction of BM haemopoietic stem cells (HPSC). The objective of this study was to investigate potential therapeutic effect of irisin, a molecule involved in adipose tissue transition, on AA mouse model. Our results showed that the concentration of irisin in serum was lower in AA patients than in healthy controls, suggesting a role of irisin in the pathogenesis of AA. In the AA mice, irisin administration prolonged the survival rate, prevented or attenuated peripheral pancytopenia, and preserved HPSC in the BM. Moreover, irisin also markedly reduced BM adipogenesis. In vitro results showed that irisin increased both cell proliferation and colony numbers of HPSC. Furthermore, our results demonstrated that irisin upregulated the expression of mitochondrial ATPase Inhibitory Factor 1 (IF1) in HPSC, inhibited the activation of mitochondrial fission protein (DRP1) and enhanced aerobic glycolysis. Taken together, our findings indicate novel roles of irisin in the pathogenesis of AA, and in the protection of HPSC through stimulation of proliferation and regulation of mitochondria function, which provides a proof-of-concept for the application of irisin in AA therapy.


Asunto(s)
Anemia Aplásica , Células Madre Hematopoyéticas , Pancitopenia , Animales , Humanos , Ratones , Anemia Aplásica/patología , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Fibronectinas/metabolismo , Fibronectinas/farmacología , Pancitopenia/metabolismo , Pancitopenia/patología , Células Madre Hematopoyéticas/efectos de los fármacos
13.
Clin Chim Acta ; 548: 117492, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37479012

RESUMEN

BACKGROUND AND AIMS: The pathophysiology of acquired aplastic anemia (aAA) is most associated with T cell mediated immune dysfunction, but the role of CD4- CD8- double negative T cells (DNTs) in pediatric patients with aAA is unclear. In this study, we aimed to investigate the proportion of TCR-αß+ DNTs in pediatric patients with aAA and correlation with the response to immunosuppressive therapy (IST). MATERIALS AND METHODS: Assessment of DNTs from peripheral blood was done by sensitive multi-color flow cytometry. The potential clinical value of TCR-αß+ DNTs was then assessed by the receiver operating characteristic (ROC) curves. RESULTS: The retrospective study evaluated 164 pediatric patients with aAA and 105 healthy donors (HD). Our data showed higher proportion of TCR-αß+ DNTs in total lymphocytes [1.04% (0.79%-1.40%) vs 0.69% (0.47%-0.87%), p < 0.001] and CD3+ T cells [1.52% (1.10%-1.96%) vs 1.10% (0.70%-1.40%), p < 0.001] in aAA compared to HD. Patients with SAA/VSAA achieving complete response (CR) after IST had a higher proportion of TCR-αß+ DNTs at initial diagnosis, than those not achieving CR for total (1.21%±0.39 vs 0.78%±0.38, p < 0.05) and CD3+ T cells (1.74%±0.53 vs 1.15%±0.59, p < 0.05). The ROC analysis showed areas under the curves (AUCs) for TCR-αß+ DNT proportion in lymphocytes and CD3+ T cells were 0.756 (cutoff value 1.33, p < 0.05) and 0.758 (cutoff value 1.38, p < 0.05), respectively. And the complete response rate was higher in TCR-αß+ DNT proportion high group than in TCR-αß+ DNT proportion low group at baseline (p < 0.001). CONCLUSION: Our observations suggest that elevated TCR-αß+ DNTs seems to play a role in the pathogenesis of aAA, and it was involve in immune response to IST.


Asunto(s)
Anemia Aplásica , Linfocitos T , Humanos , Niño , Receptores de Antígenos de Linfocitos T alfa-beta/uso terapéutico , Anemia Aplásica/tratamiento farmacológico , Anemia Aplásica/patología , Estudios Retrospectivos , Terapia de Inmunosupresión
14.
Indian J Gastroenterol ; 42(2): 279-285, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37162701

RESUMEN

Pancytopenia in children with celiac disease (CeD) is postulated to be due to nutritional deficiency such as vitamin B12, folate and copper or an autoimmune process resulting in aplastic anemia with hypoplastic marrow. In the present case series, we report the profile and explore the etiology of pancytopenia among children with CeD. There are only a few case reports of pancytopenia in children with CeD. We enrolled newly diagnosed cases of CeD and pancytopenia presenting in the celiac disease clinic over three years. Detailed evaluation was carried out for the cause of pancytopenia. We followed up on the cases for compliance and response to gluten-free diet at three months, six months and 12 months. Twenty patients were eligible for inclusion. They were divided into two groups: one with aplastic anemia with hypoplastic marrow labeled as Gp CeD-AA and the other with megaloblastic/nutritional anemia labeled as Gp CeD-MA. Patients in Gp CeD-MA presented with classical symptoms of CeD as recurrent diarrhea, abdomen distension, pallor and poor weight gain. They had none or just one transfusion requirement and had an early and complete recovery from pancytopenia. Patients in Gp CeD-AA presented with atypical symptoms such as epistaxis, short stature, fever, pallor and weakness. They had a multiple blood transfusion requirement and had delayed and partial recovery from pancytopenia. Pancytopenia is not a disease in itself but is the presentation of an underlying disease. It can occur due to various coexisting disorders in children with CeD, which can be as simple as nutritional deficiencies to as complex as an autoimmune process or malignancy. CeD should be included in the differential diagnosis of aplastic anemia as CeD and aplastic anemia both have a similar pathological process involving T cell destruction of tissues.


Asunto(s)
Anemia Aplásica , Anemia Megaloblástica , Enfermedad Celíaca , Pancitopenia , Humanos , Niño , Pancitopenia/etiología , Pancitopenia/diagnóstico , Pancitopenia/patología , Anemia Aplásica/complicaciones , Anemia Aplásica/diagnóstico , Anemia Aplásica/patología , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/diagnóstico , Palidez/complicaciones , Anemia Megaloblástica/complicaciones
15.
Bull Exp Biol Med ; 174(4): 538-543, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36899207

RESUMEN

The differentiation potential of individual clones of fibroblast CFU (CFU-F) was studied and the relative expression level of genes was analyzed in the culture of CFU-F from the bone marrow in patients with non-severe and severe forms of aplastic anemia at the onset of the disease. The differentiation potential of CFU-F clones was determined by the relative expression of marker genes using quantitative PCR. In aplastic anemia, the ratio of CFU-F clones with different differentiation potential changes, but the molecular mechanisms of this phenomenon are different in non-severe and severe aplastic anemia. In the culture of CFU-F in non-severe and severe aplastic anemia, the relative expression level of genes associated with the maintenance of the hematopoietic stem cell in the bone marrow niche changes, but the decrease in the expression of immunoregulatory genes occurs in severe form only, which may reflect differences in the pathogenesis of non-severe and severe aplastic anemia.


Asunto(s)
Anemia Aplásica , Humanos , Anemia Aplásica/genética , Anemia Aplásica/patología , Médula Ósea/patología , Células Cultivadas , Células Madre Hematopoyéticas/patología , Diferenciación Celular/genética , Gravedad del Paciente , Fibroblastos/patología , Expresión Génica/genética , Ensayo de Unidades Formadoras de Colonias
16.
Int J Mol Sci ; 24(5)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36901900

RESUMEN

Aplastic anemia (AA), a rare but potentially life-threatening disease, is a paradigm of bone marrow failure syndromes characterized by pancytopenia in the peripheral blood and hypocellularity in the bone marrow. The pathophysiology of acquired idiopathic AA is quite complex. Mesenchymal stem cells (MSCs), an important component of the bone marrow, are crucial in providing the specialized microenvironment for hematopoiesis. MSC dysfunction may result in an insufficient bone marrow and may be associated with the development of AA. In this comprehensive review, we summarized the current understanding about the involvement of MSCs in the pathogenesis of acquired idiopathic AA, along with the clinical application of MSCs for patients with the disease. The pathophysiology of AA, the major properties of MSCs, and results of MSC therapy in preclinical animal models of AA are also described. Several important issues regarding the clinical use of MSCs are discussed finally. With evolving knowledge from basic studies and clinical applications, we anticipate that more patients with the disease can benefit from the therapeutic effects of MSCs in the near future.


Asunto(s)
Anemia Aplásica , Células Madre Mesenquimatosas , Pancitopenia , Animales , Anemia Aplásica/patología , Médula Ósea/patología , Células Madre Mesenquimatosas/fisiología
17.
Clin Radiol ; 78(6): e463-e468, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36977621

RESUMEN

AIM: To determine the prospect of using machine learning with magnetic resonance imaging (MRI) to identify aplastic anaemia (AA) and myelodysplastic syndromes (MDS). MATERIALS AND METHODS: This retrospective study included patients diagnosed with AA or MDS by pathological bone marrow biopsy, who underwent pelvic MRI with the iterative decomposition of water and fat with echo asymmetry and least-squares estimation quantitation (IDEAL-IQ) between December 2016 and August 2020. Based on values of right ilium fat fraction (FF) and radiomic features extracted from T1-weighted (T1W) and IDEAL-IQ images, three machine learning algorithms including linear discriminant analysis (LDA), logistic regression (LR), and support vector machine (SVM) were used to identify AA and MDS. RESULTS: A total of 77 patients were included in the study, including 37 men and 40 women, aged 20-84 years (median age 47 years). There were 21 patients with MDS (nine men and 12 women, aged 38-84 years, median age 55 years) and 56 patients with AA (28 men and 28 women, aged 20-69 years, median age 41 years). The ilium FF of patients with AA (mean ± standard deviation [SD]: 79.23 ± 15.04%) was determined to be significantly greater compared to MDS patients (mean ± SD: 42.78 ± 30.09%, p<0.001). Selecting from the machine learning models based on ilium FF, T1W imaging and IDEAL-IQ, the IDEAL-IQ-based SVM classifier model had the best predictive ability. CONCLUSION: The combination of machine learning and IDEAL-IQ technology may enable non-invasive and accurate identification of AA and MDS.


Asunto(s)
Anemia Aplásica , Síndromes Mielodisplásicos , Masculino , Humanos , Femenino , Persona de Mediana Edad , Adulto , Anemia Aplásica/patología , Médula Ósea/patología , Estudios Retrospectivos , Síndromes Mielodisplásicos/patología , Imagen por Resonancia Magnética/métodos , Aprendizaje Automático
18.
J Autoimmun ; 136: 103026, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37001436

RESUMEN

Acquired aplastic anemia (AA) is recognized as an immune-mediated disorder resulting from active destruction of hematopoietic cells in bone marrow (BM) by effector T lymphocytes. Bulk genomic landscape analysis and transcriptomic profiling have contributed to a better understanding of the recurrent cytogenetic abnormalities and immunologic cues associated with the onset of hematopoietic destruction. However, the functional mechanistic determinants underlying the complexity of heterogeneous T lymphocyte populations as well as their correlation with clinical outcomes remain to be elucidated. To uncover dysfunctional mechanisms acting within the heterogeneous marrow-infiltrating immune environment and examine their pathogenic interplay with the hematopoietic stem/progenitor pool, we exploited single-cell mass cytometry for BM mononuclear cells of severe AA (SAA) patients pre- and post-immunosuppressive therapy, in contrast to those of healthy donors. Alignment of BM cellular composition with hematopoietic developmental trajectories revealed potential functional roles for non-canonically activated CD4+ naïve T cells in newly-diagnosed pediatric cases of SAA. Furthermore, single-cell transcriptomic profiling highlighted a population of Th17-polarized CD4+CAMK4+ naïve T cells showing activation of the IL-6/JAK3/STAT3 pathway, while gene signature dissection indicated a predisposition to proinflammatory pathogenesis. Retrospective validation from our SAA cohort of 231 patients revealed high plasma levels of IL-6 as an independent risk factor of delayed hematopoietic response to antithymocyte globulin-based immunosuppressive therapy. Thus, IL-6 warrants further investigation as a putative therapeutic target in SAA.


Asunto(s)
Anemia Aplásica , Humanos , Niño , Anemia Aplásica/genética , Anemia Aplásica/patología , Interleucina-6/genética , Estudios Retrospectivos , Células Th17 , Análisis de la Célula Individual , Janus Quinasa 3 , Factor de Transcripción STAT3/genética
19.
J Clin Oncol ; 41(1): 132-142, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36054881

RESUMEN

PURPOSE: Secondary myeloid neoplasms (sMNs) remain the most serious long-term complications in patients with aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH). However, sMNs lack specific predictors, dedicated surveillance measures, and early therapeutic interventions. PATIENTS AND METHODS: We studied a multicenter, retrospective cohort of 1,008 patients (median follow-up 8.6 years) with AA and PNH to assess clinical and molecular determinants of clonal evolution. RESULTS: Although none of the patients transplanted upfront (n = 117) developed clonal complications (either sMN or secondary PNH), the 10-year cumulative incidence of sMN in nontransplanted cases was 11.6%. In severe AA, older age at presentation and lack of response to immunosuppressive therapy were independently associated with increased risk of sMN, whereas untreated patients had the highest risk among nonsevere cases. The elapsed time from AA to sMN was 4.5 years. sMN developed in 94 patients. The 5-year overall survival reached 40% and was independently associated with bone marrow blasts at sMN onset. Myelodysplastic syndrome with high-risk phenotypes, del7/7q, and ASXL1, SETBP1, RUNX1, and RAS pathway gene mutations were the most frequent characteristics. Cross-sectional studies of clonal dynamics from baseline to evolution revealed that PIGA/human leukocyte antigen lesions decreased over time, being replaced by clones with myeloid hits. PIGA and BCOR/L1 mutation carriers had a lower risk of sMN progression, whereas myeloid driver lesions marked the group with a higher risk. CONCLUSION: The risk of sMN in AA is associated with disease severity, lack of response to treatment, and patients' age. sMNs display high-risk morphological, karyotypic, and molecular features. The landscape of acquired somatic mutations is complex and incompletely understood and should be considered with caution in medical management.


Asunto(s)
Anemia Aplásica , Hemoglobinuria Paroxística , Humanos , Anemia Aplásica/genética , Anemia Aplásica/patología , Anemia Aplásica/terapia , Hemoglobinuria Paroxística/genética , Estudios Retrospectivos , Estudios Transversales , Evolución Clonal/genética
20.
Blood ; 141(1): 72-89, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36130301

RESUMEN

Immune aplastic anemia (AA) is a severe blood disease characterized by T-lymphocyte- mediated stem cell destruction. Hematopoietic stem cell transplantation and immunosuppression are effective, but they entail costs and risks, and are not always successful. The Janus kinase (JAK) 1/2 inhibitor ruxolitinib (RUX) suppresses cytotoxic T-cell activation and inhibits cytokine production in models of graft-versus-host disease. We tested RUX in murine immune AA for potential therapeutic benefit. After infusion of lymph node (LN) cells mismatched at the major histocompatibility complex [C67BL/6 (B6)⇒CByB6F1], RUX, administered as a food additive (Rux-chow), attenuated bone marrow hypoplasia, ameliorated peripheral blood pancytopenia, preserved hematopoietic progenitors, and prevented mortality, when used either prophylactically or therapeutically. RUX suppressed the infiltration, proliferation, and activation of effector T cells in the bone marrow and mitigated Fas-mediated apoptotic destruction of target hematopoietic cells. Similar effects were obtained when Rux-chow was fed to C.B10 mice in a minor histocompatibility antigen mismatched (B6⇒C.B10) AA model. RUX only modestly suppressed lymphoid and erythroid hematopoiesis in normal and irradiated CByB6F1 mice. Our data support clinical trials of JAK/STAT inhibitors in human AA and other immune bone marrow failure syndromes.


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Pancitopenia , Ratones , Humanos , Animales , Pancitopenia/patología , Anemia Aplásica/patología , Trastornos de Fallo de la Médula Ósea/patología , Médula Ósea/patología , Enfermedades de la Médula Ósea/patología , Janus Quinasa 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA