Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.602
Filtrar
1.
3.
Heart Fail Rev ; 29(5): 1049-1063, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38985383

RESUMEN

Improvements in therapies for heart failure with preserved ejection fraction (HFpEF) are crucial for improving patient outcomes and quality of life. Although HFpEF is the predominant heart failure type among older individuals, its prognosis is often poor owing to the lack of effective therapies. The roles of the spleen and bone marrow are often overlooked in the context of HFpEF. Recent studies suggest that the spleen and bone marrow could play key roles in HFpEF, especially in relation to inflammation and immune responses. The bone marrow can increase production of certain immune cells that can migrate to the heart and contribute to disease. The spleen can contribute to immune responses that either protect or exacerbate heart failure. Extramedullary hematopoiesis in the spleen could play a crucial role in HFpEF. Increased metabolic activity in the spleen, immune cell production and mobilization to the heart, and concomitant cytokine production may occur in heart failure. This leads to systemic chronic inflammation, along with an imbalance of immune cells (macrophages) in the heart, resulting in chronic inflammation and progressive fibrosis, potentially leading to decreased cardiac function. The bone marrow and spleen are involved in altered iron metabolism and anemia, which also contribute to HFpEF. This review presents the concept of an interplay between the heart, spleen, and bone marrow in the setting of HFpEF, with a particular focus on extramedullary hematopoiesis in the spleen. The aim of this review is to discern whether the spleen can serve as a new therapeutic target for HFpEF.


Asunto(s)
Médula Ósea , Insuficiencia Cardíaca , Hematopoyesis Extramedular , Bazo , Humanos , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/metabolismo , Hematopoyesis Extramedular/fisiología , Bazo/inmunología , Bazo/metabolismo , Volumen Sistólico/fisiología , Miocardio/metabolismo , Miocardio/patología , Miocardio/inmunología , Inflamación
7.
Blood ; 144(9): 964-976, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-38728427

RESUMEN

ABSTRACT: Adult hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) hematopoietic niche, which regulates HSPC quiescence, self-renewal, and commitment in a demand-adapted manner. Although the complex BM niche is responsible for adult hematopoiesis, evidence exists for simpler, albeit functional and more accessible, extramedullary hematopoietic niches. Inspired by the anecdotal description of retroperitoneal hematopoietic masses occurring at higher frequency upon hormonal dysregulation within the adrenal gland, we hypothesized that the adult adrenal gland could be induced into a hematopoietic-supportive environment in a systematic manner, thus revealing mechanisms underlying de novo niche formation in the adult. Here, we show that upon splenectomy and hormonal stimulation, the adult adrenal gland of mice can be induced to recruit and host functional HSPCs, capable of serial transplantation, and that this phenomenon is associated with de novo formation of platelet-derived growth factor receptor α/leptin receptor (PDGFRα+/LEPR+/-)-expressing stromal nodules. We further show in CXCL12-green fluorescent protein reporter mice that adrenal glands contain a stromal population reminiscent of the CXCL12-abundant reticular cells, which compose the BM HSPC niche. Mechanistically, HSPC homing to hormonally induced adrenal glands was found dependent on the CXCR4-CXCL12 axis. Mirroring our findings in mice, we found reticular CXCL12+ cells coexpressing master niche regulator FOXC1 in primary samples from human adrenal myelolipomas, a benign tumor composed of adipose and hematopoietic tissue. Our findings reignite long-standing questions regarding hormonal regulation of hematopoiesis and provide a novel model to facilitate the study of adult-specific inducible hematopoietic niches, which may pave the way to therapeutic applications.


Asunto(s)
Glándulas Suprarrenales , Quimiocina CXCL12 , Células Madre Hematopoyéticas , Receptores CXCR4 , Nicho de Células Madre , Animales , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Quimiocina CXCL12/metabolismo , Ratones , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Ratones Endogámicos C57BL , Humanos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores de Leptina/metabolismo , Receptores de Leptina/genética , Hematopoyesis Extramedular , Esplenectomía , Ratones Transgénicos
9.
Ann Hematol ; 103(6): 1887-1896, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38581547

RESUMEN

We evaluated the impact of the genotype on clinical and hematochemical features, hepatic and cardiac iron levels, and endocrine, hepatic, and cardiovascular complications in non-transfusion-dependent (NTD) ß-thalassemia intermedia (TI) patients. Sixty patients (39.09 ± 11.11 years, 29 females) consecutively enrolled in the Myocardial Iron Overload in Thalassemia project underwent Magnetic Resonance Imaging to quantify iron overload, biventricular function parameters, and atrial areas and to detect replacement myocardial fibrosis. Three groups of patients were identified: homozygous ß+ (N = 18), heterozygous ß0ß+ (N = 22), and homozygous ß0 (N = 20). The groups were homogeneous for sex, age, splenectomy, hematochemical parameters, chelation therapy, and iron levels. The homozygous ß° genotype was associated with significantly higher biventricular end-diastolic and end-systolic volume indexes and bi-atrial area indexes. No difference was detected in biventricular ejection fractions or myocardial fibrosis. Extramedullary hematopoiesis and leg ulcers were significantly more frequent in the homozygous ß° group compared to the homozygous ß+ group. No association was detected between genotype and liver cirrhosis, hypogonadism, hypothyroidism, osteoporosis, heart failure, arrhythmias, and pulmonary hypertension. Heart remodelling related to a high cardiac output state cardiomyopathy, extramedullary hematopoiesis, and leg ulcers were more pronounced in patients with the homozygous ß° genotype compared to the other genotypes analyzed. The knowledge of the genotype can assist in the clinical management of NTD ß-TI patients.


Asunto(s)
Genotipo , Sobrecarga de Hierro , Hierro , Talasemia beta , Humanos , Talasemia beta/genética , Talasemia beta/complicaciones , Femenino , Masculino , Adulto , Persona de Mediana Edad , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/etiología , Hierro/metabolismo , Úlcera de la Pierna/etiología , Úlcera de la Pierna/genética , Hematopoyesis Extramedular/genética , Imagen por Resonancia Magnética , Miocardio/patología , Miocardio/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/complicaciones , Homocigoto
10.
Clin Med (Lond) ; 24(3): 100203, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38643827

RESUMEN

Megakaryocytes are large multilobulated precursor cells which usually reside within the bone marrow and give rise to platelets. There have been rare occurrences where they have been found in peripheral blood and extramedullary tissues in conditions where the underlying mechanisms of the bone marrow have been affected. This case report discusses an unusual presentation of a man with myelofibrosis who was found to have megakaryocytes in his ascitic fluid. We have highlighted the images showing utility of combination of traditional staining methods and immunohistochemistry in combating this diagnostic dilemma.


Asunto(s)
Hematopoyesis Extramedular , Megacariocitos , Mielofibrosis Primaria , Humanos , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/patología , Masculino , Megacariocitos/patología , Persona de Mediana Edad
11.
Am J Hematol ; 99(7): 1326-1337, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38646919

RESUMEN

Emerging evidence shows the crucial role of inflammation (particularly NF-κB pathway) in the development and progression of myelofibrosis (MF), becoming a promising therapeutic target. Furthermore, tailoring treatment with currently available JAK inhibitors (such as ruxolitinib or fedratinib) does not modify the natural history of the disease and has important limitations, including cytopenias. Since recent studies have highlighted the role of miR-146a, a negative regulator of the NF-κB pathway, in the pathogenesis of MF; here we used miR-146a-/- (KO) mice, a MF-like model lacking driver mutations, to investigate whether pharmacological inhibition of JAK/STAT and/or NF-κB pathways may reverse the myelofibrotic phenotype of these mice. Specifically, we tested the JAK1/2 inhibitor, ruxolitinib; the NF-κB inhibitor via IKKα/ß, BMS-345541; both inhibitors in combination; or a dual inhibitor of both pathways (JAK2/IRAK1), pacritinib. Although all treatments decreased spleen size and partially recovered its architecture, only NF-κB inhibition, either using BMS-345541 (alone or in combination) or pacritinib, resulted in a reduction of extramedullary hematopoiesis, bone marrow (BM) fibrosis and osteosclerosis, along with an attenuation of the exacerbated inflammatory state (via IL-1ß and TNFα). However, although dual inhibitor improved anemia and reversed thrombocytopenia, the combined therapy worsened anemia by inducing BM hypoplasia. Both therapeutic options reduced NF-κB and JAK/STAT signaling in a context of JAK2V617F-driven clonal hematopoiesis. Additionally, combined treatment reduced both COL1A1 and IL-6 production in an in vitro model mimicking JAK2-driven fibrosis. In conclusion, NF-κB inhibition reduces, in vitro and in vivo, disease burden and BM fibrosis, which could provide benefits in myelofibrosis patients.


Asunto(s)
Ratones Noqueados , MicroARNs , FN-kappa B , Nitrilos , Mielofibrosis Primaria , Pirazoles , Pirimidinas , Animales , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , MicroARNs/genética , Ratones , FN-kappa B/metabolismo , Nitrilos/uso terapéutico , Nitrilos/farmacología , Pirimidinas/uso terapéutico , Pirimidinas/farmacología , Pirazoles/uso terapéutico , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Hematopoyesis Extramedular/efectos de los fármacos
13.
Exp Mol Med ; 56(3): 549-558, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38443597

RESUMEN

Hematopoiesis can occur outside of the bone marrow during inflammatory stress to increase the production of primarily myeloid cells at extramedullary sites; this process is known as extramedullary hematopoiesis (EMH). As observed in a broad range of hematologic and nonhematologic diseases, EMH is now recognized for its important contributions to solid tumor pathology and prognosis. To initiate EMH, hematopoietic stem cells (HSCs) are mobilized from the bone marrow into the circulation and to extramedullary sites such as the spleen and liver. At these sites, HSCs primarily produce a pathological subset of myeloid cells that contributes to tumor pathology. The EMH HSC niche, which is distinct from the bone marrow HSC niche, is beginning to be characterized. The important cytokines that likely contribute to initiating and maintaining the EMH niche are KIT ligands, CXCL12, G-CSF, IL-1 family members, LIF, TNFα, and CXCR2. Further study of the role of EMH may offer valuable insights into emergency hematopoiesis and therapeutic approaches against cancer. Exciting future directions for the study of EMH include identifying common and distinct EMH mechanisms in cancer, infectious diseases, and chronic autoimmune diseases to control these conditions.


Asunto(s)
Hematopoyesis Extramedular , Neoplasias , Humanos , Hematopoyesis , Células Madre Hematopoyéticas , Médula Ósea , Enfermedad Crónica
14.
Exp Hematol ; 134: 104177, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38336135

RESUMEN

Emerging evidence implicates the epithelial-mesenchymal transition transcription factor Zeb1 as a critical regulator of hematopoietic stem cell (HSC) differentiation. Whether Zeb1 regulates long-term maintenance of HSC function remains an open question. Using an inducible Mx-1-Cre mouse model that deletes conditional Zeb1 alleles in the adult hematopoietic system, we found that mice engineered to be deficient in Zeb1 for 32 weeks displayed expanded immunophenotypically defined adult HSCs and multipotent progenitors associated with increased abundance of lineage-biased/balanced HSC subsets and augmented cell survival characteristics. During hematopoietic differentiation, persistent Zeb1 loss increased B cells in the bone marrow and spleen and decreased monocyte generation in the peripheral blood. In competitive transplantation experiments, we found that HSCs from adult mice with long-term Zeb1 deletion displayed a cell autonomous defect in multilineage differentiation capacity. Long-term Zeb1 loss perturbed extramedullary hematopoiesis characterized by increased splenic weight and a paradoxical reduction in splenic cellularity that was accompanied by HSC exhaustion, lineage-specific defects, and an accumulation of aberrant, preleukemic like c-kit+CD16/32+ progenitors. Loss of Zeb1 for up to 42 weeks can lead to progressive splenomegaly and an accumulation of Gr-1+Mac-1+ cells, further supporting the notion that long-term expression of Zeb1 suppresses preleukemic activity. Thus, sustained Zeb1 deletion disrupts HSC functionality in vivo and impairs regulation of extramedullary hematopoiesis with potential implications for tumor suppressor functions of Zeb1 in myeloid neoplasms.


Asunto(s)
Hematopoyesis Extramedular , Células Madre Hematopoyéticas , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Animales , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Ratones , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Hematopoyesis Extramedular/genética , Diferenciación Celular , Ratones Noqueados , Bazo/metabolismo , Bazo/patología , Bazo/citología , Células Madre Adultas/metabolismo , Linaje de la Célula
15.
J Int Med Res ; 52(1): 3000605231221088, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38190843

RESUMEN

Extramedullary hematopoiesis (EMH) is a rare condition characterized by proliferation of hematopoietic stem cells outside the bone marrow, usually as a compensatory response to hematological disease. Although EMH primarily occurs in the liver and spleen, it can manifest in atypical locations, such as the mediastinum. We herein describe an asymptomatic 66-year-old man with incidentally discovered posterior mediastinal EMH. A 28- × 32-mm mass was detected during a routine examination. Laboratory findings were within normal limits. Computed tomography revealed a well-defined enhancing mass with a density of 60 Hounsfield units, suggestive of a neurogenic tumor. Surgical resection confirmed EMH, characterized by megakaryocytes and hematopoietic precursors. The patient recovered smoothly and was discharged 5 days postoperatively. Accurate preoperative diagnosis of EMH is challenging, as illustrated by this case. Although typically associated with anemia or hematological abnormalities, EMH can present without such signs. Surgical resection and histopathological examination are essential for diagnosis. This case emphasizes the diagnostic complexity of posterior mediastinal EMH, even in patients without overt hematological disorders. Posterior mediastinal EMH is exceedingly rare and diagnostically demanding. A high index of suspicion and histological tissue analysis are crucial for optimal management. Video-assisted thoracoscopic surgery enables both diagnosis and treatment through mass excision.


Asunto(s)
Enfermedades Hematológicas , Hematopoyesis Extramedular , Masculino , Humanos , Anciano , Mediastino/diagnóstico por imagen , Mediastino/cirugía , Bazo , Cirugía Torácica Asistida por Video
18.
J Am Vet Med Assoc ; 262(4): 1-3, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38266389

RESUMEN

OBJECTIVE: Synovial extramedullary hematopoiesis is a rarely reported condition in humans and, to date, has never been reported in canines. This case report describes the clinical presentation, diagnostic work-up, treatment, and outcome of a canine case confirmed to have hematopoietic tissue within multiple joints. ANIMAL: A client-owned canine. CLINICAL PRESENTATION, PROGRESSION, AND PROCEDURES: The clinical presentation was most consistent with immune-mediated polyarthritis, and arthrocentesis was performed in multiple joints for cytological evaluation and culture. Cytology revealed evidence of extramedullary hematopoiesis, and shortly thereafter the dog was diagnosed with immune-mediated hemolytic anemia and thrombocytopenia. TREATMENT AND OUTCOME: Pregabalin, prednisolone, clopidogrel, and cyclosporine were started, and after several recheck appointments and dose adjustments, the dog's clinical signs resolved for all conditions. CLINICAL RELEVANCE: Unusual sites of extramedullary hematopoietic tissue may result in a clinical presentation for which more traditional etiologies and differentials are not applicable.


Asunto(s)
Anemia , Enfermedades de los Perros , Hematopoyesis Extramedular , Humanos , Perros , Animales , Médula Ósea , Anemia/veterinaria , Enfermedades de los Perros/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA