Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 13(21): 23981-24016, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34762598

RESUMEN

This study addresses the potential to reverse age-associated morbidity by establishing methods to restore the aged hematopoietic system. Parabiotic animal models indicated that young secretome could restore aged tissues, leading us to establish a heterochronic transwell system with aged mobilized peripheral blood (MPB), co-cultured with young MPB or umbilical cord blood (UCB) cells. Functional studies and omics approaches indicate that the miRNA cargo of microvesicles (MVs) restores the aged hematopoietic system. The in vitro findings were validated in immune deficient (NSG) mice carrying an aged hematopoietic system, improving aged hallmarks such as increased lymphoid:myeloid ratio, decreased inflammation and cellular senescence. Elevated MYC and E2F pathways, and decreased p53 were key to hematopoietic restoration. These processes require four restorative miRs that target the genes for transcription/differentiation, namely PAX and phosphatase PPMIF. These miRs when introduced in aged cells were sufficient to restore the aged hematopoietic system in NSG mice. The aged MPBs were the drivers of their own restoration, as evidenced by the changes from distinct baseline miR profiles in MPBs and UCB to comparable expressions after exposure to aged MPBs. Restorative natural killer cells eliminated dormant breast cancer cells in vivo, indicating the broad relevance of this cellular paradigm - preventing and reversing age-associated disorders such as clearance of early malignancies and enhanced responses to vaccine and infection.


Asunto(s)
Células de la Médula Ósea , Micropartículas Derivadas de Células , Senescencia Celular/fisiología , Hematopoyesis/fisiología , Adulto , Anciano , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/fisiología , Femenino , Sangre Fetal/citología , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Secretoma , Adulto Joven
2.
Stem Cell Rev Rep ; 17(6): 2178-2192, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34410592

RESUMEN

Mesenchymal stem cells (MSCs) can become dysfunctional in patients with hematological disorders. An unanswered question is whether age-linked disruption of the bone marrow (BM) microenvironment is secondary to hematological dysfunction or vice versa. We therefore studied MSC function in patients with different hematological disorders and found decreased MHC-II except from one sample with acute myeloid leukemia (AML). The patients' MSCs were able to exert veto properties except for AML MSCs. While the expression of MHC-II appeared to be irrelevant to the immune licensing of MSCs, AML MSCs lost their ability to differentiate upon contact and rather, continued to proliferate, forming foci-like structures. We performed a retrospective study that indicated a significant increase in MSCs, based on phenotype, for patients with BM fibrosis. This suggests a role for MSCs in patients transitioning to leukemia. NFĸB was important to MSC function and was shown to be a potential target to sensitize leukemic CD34+/CD38- cells to azacitidine. This correlated with their lack of allogeneic stimulation. This study identified NFĸB as a potential target for combination therapy to treat leukemia stem cells and showed that understanding MSC biology and immune response could be key in determining how the aging BM might support leukemia. More importantly, we show how MSCs might be involved in transitioning the high risk patient with hematological disorder to AML.


Asunto(s)
Neoplasias Hematológicas , Células Madre Mesenquimatosas , Células de la Médula Ósea , Proliferación Celular , Neoplasias Hematológicas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Estudios Retrospectivos , Microambiente Tumoral
3.
Methods Mol Biol ; 2224: 195-202, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606216

RESUMEN

Mice with human hematopoietic system have become critical for research and preclinical studies. Mice with patient-derived xenografts of different tumors exist without human immune system. Answers can be addressed with the same immunodeficient mice that are chimeric for the human hemato-lymphoid system (humanized mice). The growing field of immune-oncology could benefit from preclinical studies with the humanized mice. Other fields will also benefit such as studies of infectious disease, regenerative medicine, organ transplant, and allergies. Here, we describe the method to humanize immune-deficient mice with human CD34+ hematopoietic cells.


Asunto(s)
Sistema Inmunológico/inmunología , Síndromes de Inmunodeficiencia/inmunología , Adulto , Animales , Antígenos CD34/inmunología , Modelos Animales de Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad
4.
World J Stem Cells ; 12(11): 1341-1353, 2020 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-33312402

RESUMEN

The potential clinical and economic impact of mesenchymal stem cell (MSC) therapy is immense. MSCs act through multiple pathways: (1) as "trophic" cells, secreting various factors that are immunomodulatory, anti-inflammatory, anti-apoptotic, proangiogenic, proliferative, and chemoattractive; (2) in conjunction with cells native to the tissue they reside in to enhance differentiation of surrounding cells to facilitate tissue regrowth. Researchers have developed methods for the extraction and expansion of MSCs from animal and human tissues. While many sources of MSCs exist, including adipose tissue and iliac crest bone graft, compact bone (CB) MSCs have shown great potential for use in orthopaedic surgery. CB MSCs exert powerful immunomodulatory effects in addition to demonstrating excellent regenerative capacity for use in filling boney defects. CB MSCs have been shown to have enhanced response to hypoxic conditions when compared with other forms of MSCs. More work is needed to continue to characterize the potential applications for CB MSCs in orthopaedic trauma.

5.
Cancer Lett ; 488: 9-17, 2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32479768

RESUMEN

Breast cancer (BC) remains a clinical challenge despite improved treatments and public awareness to ensure early diagnosis. A major issue is the ability of BC cells (BCCs) to survive as dormant cancer cells in the bone marrow (BM), resulting in the cancer surviving for decades with the potential to resurge as metastatic cancer. The experimental evidence indicates similarity between dormant BCCs and other stem cells, resulting in the preponderance of data to show dormant BCCs being cancer stem cells (CSCs). The BM niche and their secretome support BCC dormancy. Lacking in the literature is a comprehensive research to describe how the hypoxic environment within the BM may influence the behavior of BCCs. This information is relevant to understand the prognosis of BC in young and aged individuals whose oxygen levels differ in BM. This review discusses the changing information on vascularity in different regions of the BM and the impact on endogenous hematopoietic stem cells (HSCs). This review highlights the necessary information to provide insights on vascularity of different BM regions on the behavior of BCCs, in particular a dormant phase. For instance, how the transcription factor HIF1-α (hypoxia-inducible factor 1 alpha), functioning as first responder under hypoxic conditions, affects the expression of specific gene networks involved in energy metabolism, cell survival, tumor invasion and angiogenesis. This enables cell fate transition and facilitates tumor heterogeneity, which in turn favors tumor progression and resistance to anticancer treatments Thus, HIF1-α could be a potential target for cancer treatment. This review describes epigenetic mechanisms involved in hypoxic responses during cancer dormancy in the bone marrow. The varied hypoxic environment in the BM is relevant to understand the complex process of the aging bone marrow for insights on breast cancer outcome between the young and aged.


Asunto(s)
Neoplasias de la Médula Ósea/secundario , Neoplasias de la Mama/patología , Hipoxia de la Célula/fisiología , Células Madre Neoplásicas/patología , Microambiente Tumoral/fisiología , Animales , Médula Ósea/patología , Femenino , Humanos
6.
J Immunol ; 204(4): 879-891, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31924647

RESUMEN

Hematopoiesis is tightly regulated by the bone marrow (BM) niche. The niche is robust, allowing for the return of hematopoietic homeostasis after insults such as infection. Hematopoiesis is partly regulated by soluble factors, such as neuropeptides, substance P (SP), and neurokinin A (NK-A), which mediate hematopoietic stimulation and inhibition, respectively. SP and NK-A are derived from the Tac1 gene that is alternately spliced into four variants. The hematopoietic effects of SP and NK-A are mostly mediated via BM stroma. Array analyses with 2400 genes indicated distinct changes in SP-stimulated BM stroma. Computational analyses indicated networks of genes with hematopoietic regulation. Included among these networks is the high-mobility group box 1 gene (HMGB1), a nonhistone chromatin-associated protein. Validation studies indicated that NK-A could reverse SP-mediated HMGB1 decrease. Long-term culture-initiating cell assay, with or without NK-A receptor antagonist (NK2), showed a suppressive effect of HMGB1 on hematopoietic progenitors and increase in long-term culture-initiating cell assay cells (primitive hematopoietic cells). These effects occurred partly through NK-A. NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Although the effects on myeloid lineage were suppressed, the results suggested a more complex effect on the lymphoid lineage. Clonogenic assay for CFU- granulocyte-monocyte suggested that HMGB1 may be required to prevent hematopoietic stem cell exhaustion to ensure immune homeostasis. In summary, this study showed how HMGB1 is linked to SP and NK-A to protect the most primitive hematopoietic cell and also to maintain immune/hematopoietic homeostasis.


Asunto(s)
Proteína HMGB1/metabolismo , Hematopoyesis/genética , Neuroinmunomodulación/genética , Neuroquinina A/metabolismo , Sustancia P/metabolismo , Adolescente , Adulto , Empalme Alternativo , Animales , Benzamidas/farmacología , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/inmunología , Células HEK293 , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Neuroinmunomodulación/inmunología , Neuroquinina A/antagonistas & inhibidores , Análisis de Secuencia por Matrices de Oligonucleótidos , Piperidinas/farmacología , Cultivo Primario de Células , Taquicininas/genética , Quimera por Trasplante , Adulto Joven
7.
Cell Death Dis ; 10(2): 59, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683851

RESUMEN

Breast cancer (BC) cells (BCCs) can retain cellular quiescence for decades, a phenomenon referred to as dormancy. BCCs show preference for the bone marrow (BM) where they can remain dormant for decades. Targeting BCCs within the BM is a challenge since the dormant BCCs reside within BM stroma, also residence for hematopoietic stem cells (HSCs). Dormant BCCs could behave as cancer stem cells (CSCs). The CSCs and HSCs are similar by function and also, by commonly expressed genes. The method by which dormant BCCs transition into clinically metastatic cells remains unclear. This study tested the hypothesis that macrophages (MΦs) within BM stroma, facilitates dormancy or reverse this state into metastatic cells. MΦs exhibiting an M2 phenotype constitute ~10% of cultured BM stroma. The M2 MΦs form gap junctional intercellular communication (GJIC) with CSCs, resulting in cycling quiescence, reduced proliferation and carboplatin resistance. In contrast, MΦs expressing the M1 phenotype reversed BC dormancy. Activation of M2a MΦs via the toll-like receptor 4 (TLR4) switched to M1 phenotype. The switch can occur by direct activation of M2a MΦs, or indirectly through activation of mesenchymal stem cells. M1 MΦ-derived exosomes activated NFкB to reverse quiescent BCCs to cycling cells. Using an in vivo model of BC dormancy, injected Mi MOs sensitized BCCs to carboplatin and increased host survival. In summary, we have shown how BM stromal MΦs, through exosomes, regulate the behavior of BCCs, by either inducing or reversing dormancy.


Asunto(s)
Médula Ósea/patología , Neoplasias de la Mama/patología , Comunicación Celular , Exosomas/metabolismo , Macrófagos/metabolismo , Células Madre Neoplásicas/metabolismo , Adolescente , Adulto , Animales , Neoplasias de la Mama/tratamiento farmacológico , Carboplatino/uso terapéutico , Células Cultivadas , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Femenino , Uniones Comunicantes , Xenoinjertos , Humanos , Macrófagos/clasificación , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptor Toll-Like 4/metabolismo , Adulto Joven
8.
Adv Exp Med Biol ; 1201: 93-108, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31898783

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent cells that can self-renew and differentiate into cells of all germ layers. MSCs can be easily attracted to the site of tissue insult with high levels of inflammatory mediators. The general ability of MSCs to migrate at the sites of tissue injury suggested an innate ability for these cells to be involved in baseline tissue repair. The bone marrow is one of the primary sources of MSCs, though they can be ubiquitous. An attractive property of MSCs for clinical application is their ability to cross allogeneic barrier. However, alone, MSCs are not immune suppressive cells. Rather, they can be licensed by the tissue microenvironment to become immune suppressor cells. Immune suppressor functions of MSCs include those that blunt cytotoxicity of natural killer cells, suppression of T-cell proliferation, and "veto" function. MSCs, as third-party cells, suppress the immune response that generally recapitulates graft-versus-host disease (GvHD) responses. Based on the plastic functions of MSCs, these cells have dominated the field of cell-based therapies, such as anti-inflammatory and drug delivery. Here, we focus on the potential use of MSC for immunological disorders such as Crohn's disease and GvHD.


Asunto(s)
Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/terapia , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Humanos , Enfermedades del Sistema Inmune/patología , Células Asesinas Naturales/inmunología , Células Madre Mesenquimatosas/citología
9.
Biochimie ; 155: 92-103, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29859990

RESUMEN

The secretome produced by cells within the bone marrow is significant to homeostasis. The bone marrow, a well-studied organ, has multiple niches with distinct roles for supporting stem cell functions. Thus, an understanding of mediators involved in the regulation of stem cells could serve as a model for clinical problems and solutions such as tissue repair and regeneration. The exosome secretome of bone marrow stem cells is a developing area of research with respect to the regenerative potential by bone marrow cell, particularly the mesenchymal stem cells. The bone marrow niche regulates endogenous processes such as hematopoiesis but could also support the survival of tumors such as facilitating the cancer stem cells to exist in dormancy for decades. The bone marrow-derived secretome will be critical to future development of therapeutic strategies for oncologic diseases, in addition to regenerative medicine. This article discusses the importance for parallel studies to determine how the same secretome may compromise safety during the use of stem cells in regenerative medicine.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Nicho de Células Madre , Animales , Médula Ósea/patología , Supervivencia Celular , Humanos , Células Madre Mesenquimatosas/patología , Neoplasias/patología
10.
Adv Exp Med Biol ; 1056: 137-152, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29754179

RESUMEN

Human aging is an inevitable and complex phenomenon characterized by a progressive, gradual degradation of physiological and cellular processes that leads from vulnerability to death. Mammalian somatic cells display limited proliferative properties in vitro that results in a process of permanent cell cycle arrest commonly known as senescence. Events leading to cellular senescence are complex but may be due to the increase in tumor suppressor genes, caused by lifetime somatic mutations. Cumulative mutation leaves an imprint on the genome of the cell, an important risk factor for the occurrence of cancer. Adults over the age of 65+ are vulnerable to age related diseases such as cancers but such changes may begin at middle age. MicroRNAs (miRNAs), which are small non-coding RNA, can regulate cancer progression, recurrence and metastasis. This chapter discusses the role of miRNA in tumor microenvironment, consequent to aging.


Asunto(s)
Envejecimiento/genética , Senescencia Celular/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias/genética , ARN Neoplásico/genética , Microambiente Tumoral/genética , Anciano , Comunicación Celular , Progresión de la Enfermedad , Epigénesis Genética/genética , Humanos , Inflamación/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/uso terapéutico , Metástasis de la Neoplasia , Neoplasias/patología , Neoplasias/terapia , Oncogenes/genética , ARN Neoplásico/antagonistas & inhibidores , Recurrencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA