Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Front Immunol ; 13: 1005554, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36311725

RESUMEN

Functional impairment of the bone marrow (BM) niche has been suggested as a major reason for prolonged cytopenia and secondary graft failure after allogeneic hematopoietic cell transplantation (alloHCT). Because mesenchymal stromal cells (MSCs) serve as multipotent progenitors for several niche components in the BM, they might play a key role in this process. We used collagenase digested trephine biopsies to directly quantify MSCs in 73 patients before (n = 18) and/or after alloHCT (n = 65). For the first time, we demonstrate that acute graft-versus-host disease (aGvHD, n = 39) is associated with a significant decrease in MSC numbers. MSC reduction can be observed even before the clinical onset of aGvHD (n = 10). Assessing MSCs instantly after biopsy collection revealed phenotypic and functional differences depending on the occurrence of aGvHD. These differences vanished during ex vivo expansion. The MSC endotypes observed revealed an enhanced population of donor-derived classical dendritic cells type 1 and alloreactive T cells as the causing agent for compartmental inflammation and MSC damage before clinical onset of aGvHD was ascertained. In conclusion, MSCs endotypes may constitute a predisposing conductor of alloreactivity after alloHCT preceding the clinical diagnosis of aGvHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Enfermedad Injerto contra Huésped/diagnóstico , Médula Ósea/patología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Mesenquimatosas/metabolismo
2.
Nat Commun ; 13(1): 4485, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35918329

RESUMEN

The benefit of molecularly-informed therapies in cancer of unknown primary (CUP) is unclear. Here, we use comprehensive molecular characterization by whole genome/exome, transcriptome and methylome analysis in 70 CUP patients to reveal substantial mutational heterogeneity with TP53, MUC16, KRAS, LRP1B and CSMD3 being the most frequently mutated known cancer-related genes. The most common fusion partner is FGFR2, the most common focal homozygous deletion affects CDKN2A. 56/70 (80%) patients receive genomics-based treatment recommendations which are applied in 20/56 (36%) cases. Transcriptome and methylome data provide evidence for the underlying entity in 62/70 (89%) cases. Germline analysis reveals five (likely) pathogenic mutations in five patients. Recommended off-label therapies translate into a mean PFS ratio of 3.6 with a median PFS1 of 2.9 months (17 patients) and a median PFS2 of 7.8 months (20 patients). Our data emphasize the clinical value of molecular analysis and underline the need for innovative, mechanism-based clinical trials.


Asunto(s)
Neoplasias Primarias Desconocidas , Epigenómica , Genómica , Homocigoto , Humanos , Mutación , Neoplasias Primarias Desconocidas/tratamiento farmacológico , Neoplasias Primarias Desconocidas/genética , Eliminación de Secuencia
3.
Cytotherapy ; 22(1): 21-26, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31883948

RESUMEN

Isolation of mesenchymal stromal cells (MSCs) from pretreated, hematologic patients is challenging. Especially after allogeneic hematopoietic cell transplantation (HCT), standard protocols using bone marrow aspirates fail to reliably recover sufficient cell numbers. Because MSCs are considered to contribute to processes that mainly affect the outcome after transplantation, such as an efficient lymphohematopoietic recovery, extent of graft-versus-host disease as well as the occurrence of leukemic relapse, it is of great clinical relevance to investigate MSC function in this context. Previous studies showed that MSCs can be isolated by collagenase digestion of large bone fragments of hematologically healthy patients undergoing hip replacement or knee surgeries. We have now further developed this procedure for the isolation of MSCs from hematologic patients after allogeneic HCT by using trephine biopsy specimens obtained during routine examinations. Comparison of aspirates and trephine biopsy specimens from patients after allogeneic HCT revealed a significantly higher frequency of clonogenic MSCs (colony-forming unit-fibroblast [CFU-F]) in trephine biopsy specimens (mean, 289.8 ± standard deviation 322.5 CFU-F colonies/1 × 106 total nucleated cells versus 4.2 ± 9.9; P < 0.0001). Subsequent expansion of functional MSCs isolated from trephine biopsy specimen was more robust and led to a significantly higher yield compared with control samples expanded from aspirates (median, 1.6 × 106; range, 0-2.3 × 107 P0 MSCs versus 5.4 × 104; range, 0-8.9 × 106; P < 0.0001). Using trephine biopsy specimens as MSC source facilitates the investigation of various clinical questions.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia/terapia , Células Madre Mesenquimatosas/citología , Adulto , Anciano , Biopsia , Médula Ósea , Colagenasas/farmacología , Femenino , Enfermedad Injerto contra Huésped/patología , Humanos , Masculino , Persona de Mediana Edad , Células Tumorales Cultivadas , Adulto Joven
5.
Adv Sci (Weinh) ; 6(8): 1802104, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-31016116

RESUMEN

Human bone marrow mesenchymal stromal cells (MSCs) are used in clinical trials for the treatment of systemic inflammatory diseases due to their regenerative and immunomodulatory properties. However, intravenous administration of MSCs is hampered by cell trapping within the pulmonary capillary networks. Here, it is hypothesized that traditional 2D plastic-adherent cell expansion fails to result in appropriate morphorheological properties required for successful cell circulation. To address this issue, a method to culture MSCs in nonadherent 3D spheroids (mesenspheres) is adapted. The biological properties of mesensphere-cultured MSCs remain identical to conventional 2D cultures. However, morphorheological analyses reveal a smaller size and lower stiffness of mesensphere-derived MSCs compared to plastic-adherent MSCs, measured using real-time deformability cytometry and atomic force microscopy. These properties result in an increased ability to pass through microconstrictions in an ex vivo microcirculation assay. This ability is confirmed in vivo by comparison of cell accumulation in various organ capillary networks after intravenous injection of both types of MSCs in mouse. The findings generally identify cellular morphorheological properties as attractive targets for improving microcirculation and specifically suggest mesensphere culture as a promising approach for optimized MSC-based therapies.

6.
Int J Cancer ; 136(1): 44-54, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24806942

RESUMEN

We investigated whether breast tumor cells can modulate the function of mesenchymal stromal cells (MSCs) with a special emphasis on their chemoattractive activity towards hematopoietic stem and progenitor cells (HSPCs). Primary MSCs as well as a MSC line (SCP-1) were cocultured with primary breast cancer cells, MCF-7, MDA-MB231 breast carcinoma or MCF-10A non-malignant breast epithelial cells or their conditioned medium. In addition, the frequency of circulating clonogenic hematopoietic progenitors was determined in 78 patients with breast cancer and compared with healthy controls. Gene expression analysis of SCP-1 cells cultured with MCF-7 medium revealed CXCL12 (SDF-1) as one of the most significantly downregulated genes. Supernatant from both MCF-7 and MDA-MB231 reduced the CXCL12 promoter activity in SCP-1 cells to 77% and 47%, respectively. Moreover, the CXCL12 mRNA and protein levels were significantly reduced. As functional consequence of lower CXCL12 levels, we detected a decreased trans-well migration of HSPCs towards MSC/tumor cell cocultures or conditioned medium. The specificity of this effect was confirmed by blocking studies with the CXCR4 antagonist AMD3100. Downregulation of SP1 and increased miR-23a levels in MSCs after contact with tumor cell medium as well as enhanced TGFß1 expression were identified as potential molecular regulators of CXCL12 activity in MSCs. Moreover, we observed a significantly higher frequency of circulating colony-forming hematopoietic progenitors in patients with breast cancer compared with healthy controls. Our in vitro results propose a potential new mechanism by which disseminated tumor cells in the bone marrow may interfere with hematopoiesis by modulating CXCL12 in protected niches.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quimiocina CXCL12/metabolismo , Células Madre Mesenquimatosas/fisiología , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Quimiocina CXCL12/genética , Quimiotaxis , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Hematopoyesis , Humanos , Células MCF-7 , MicroARNs/genética , MicroARNs/metabolismo , Células Neoplásicas Circulantes/patología , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
7.
Haematologica ; 98(11): 1677-85, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23716561

RESUMEN

The contribution of the bone marrow microenvironment in myelodysplastic syndrome is controversial. We therefore analyzed the functional properties of primary mesenchymal stromal cells from patients with myelodysplastic syndrome in the presence or absence of lenalidomide. Compared to healthy controls, clonality and growth were reduced across all disease stages. Furthermore, differentiation defects and particular expression of adhesion and cell surface molecules (e.g. CD166, CD29, CD146) were detected. Interestingly, the levels of stromal derived factor 1-alpha in patients' cells culture supernatants were almost 2-fold lower (P<0.01) than those in controls and this was paralleled by a reduced induction of migration of CD34(+) hematopoietic cells. Co-cultures of mesenchymal stromal cells from patients with CD34(+) cells from healthy donors resulted in reduced numbers of cobblestone area-forming cells and fewer colony-forming units. Exposure of stromal cells from patients and controls to lenalidomide led to a further reduction of stromal derived factor 1-alpha secretion and cobblestone area formation, respectively. Moreover, lenalidomide pretreatment of mesenchymal stromal cells from patients with low but not high-risk myelodysplastic syndrome was able to rescue impaired erythroid and myeloid colony formation of early hematopoietic progenitors. In conclusion, our analyses support the notion that the stromal microenvironment is involved in the pathophysiology of myelodysplastic syndrome thus representing a potential target for therapeutic interventions.


Asunto(s)
Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/patología , Talidomida/análogos & derivados , Anciano , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Estudios de Cohortes , Femenino , Humanos , Lenalidomida , Masculino , Persona de Mediana Edad , Talidomida/farmacología , Talidomida/uso terapéutico
8.
Cytotherapy ; 12(1): 17-30, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19878082

RESUMEN

BACKGROUND AIMS: Human adult bone marrow (BM)-derived mesenchymal stromal cells (hMSC) are reported to break germ layer commitment and differentiate into cells expressing neuroectodermal properties. Although it is of pivotal interest for cell replacement therapies for neurologic disorders, no data exist on the influence of the donor's age on this multipotent differentiation behavior. METHODS: We evaluated various epigenetic neuroectodermal conversion protocols in adult hMSC derived from older donors (>45 versus 18-35 years of age) using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and immunocytochemistry. The protocols included single- and multi-step conversion-differentiation protocols combined with co-culture techniques. Furthermore, the age dependency of mesodermal differentiation potential and cell senescence were investigated. RESULTS: The neuroectodermal differentiation potential of hMSC derived from old donors was completely lost, with no cells showing mature neuroectodermal phenotypes using single- and multi-step conversion-differentiation protocols and no improvement of neurogenesis by various co-culture conditions. Comparison of young versus old donor-derived hMSC showed fewer cells expressing early neuroectodermal marker proteins in the latter samples. qRT-PCR showed reduced expression of the proliferation marker KI67 and the neuroectodermal gene NES (nestin) in old donor-derived cells compared with young donor hMSC. Telomere length analysis showed no general cell aging. CONCLUSIONS: Our data provide evidence that only young donor-derived hMSC can be epigenetically differentiated in vitro into neuroectodermal cells, pointing towards senescence of multipotentiality of old donor-derived hMSC. There is thus an urgent need to develop better protocols for successful neuroectodermal differentiation of hMSC from old individuals as a prerequisite for autologous cell replacement strategies for neurologic diseases in elderly patients.


Asunto(s)
Diferenciación Celular/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Placa Neural/fisiología , Células del Estroma/fisiología , Células del Estroma/trasplante , Adolescente , Adulto , Factores de Edad , Anciano , Envejecimiento/fisiología , Biomarcadores/metabolismo , Proliferación Celular , Senescencia Celular/fisiología , Técnicas de Cocultivo , Contraindicaciones , Femenino , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/citología , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Nestina , Placa Neural/citología , Células del Estroma/citología , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA