Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36142235

RESUMEN

Chronic Myeloid Leukemia (CML) originates in a leukemic stem cell that resides in the bone marrow microenvironment, where they coexist with cellular and non-cellular elements. The vascular microenvironment has been identified as an important element in CML development since an increase in the vascularization has been suggested to be related with poor prognosis; also, using murine models, it has been reported that bone marrow endothelium can regulate the quiescence and proliferation of leukemic stem and progenitor cells. This observation, however, has not been evaluated in primary human cells. In this report, we used a co-culture of primitive (progenitor and stem) CML cells with endothelial colony forming cells (ECFC) as an in vitro model to evaluate the effects of the vascular microenvironment in the leukemic hematopoiesis. Our results show that this interaction allows the in vitro maintenance of primitive CML cells through an inflammatory microenvironment able to regulate the proliferation of progenitor cells and the permanence in a quiescent state of leukemic stem cells.


Asunto(s)
Células Endoteliales , Leucemia Mielógena Crónica BCR-ABL Positiva , Animales , Médula Ósea , Enfermedad Crónica , Hematopoyesis , Humanos , Ratones , Células Madre Neoplásicas , Microambiente Tumoral
2.
Cancer Control ; 28: 10732748211038735, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34565215

RESUMEN

Since the second half of the 20th century, our knowledge about the biology of cancer has made extraordinary progress. Today, we understand cancer at the genomic and epigenomic levels, and we have identified the cell that starts neoplastic transformation and characterized the mechanisms for the invasion of other tissues. This knowledge has allowed novel drugs to be designed that act on specific molecular targets, the immune system to be trained and manipulated to increase its efficiency, and ever more effective therapeutic strategies to be developed. Nevertheless, we are still far from winning the war against cancer, and thus biomedical research in oncology must continue to be a global priority. Likewise, there is a need to reduce unequal access to medical services and improve prevention programs, especially in countries with a low human development index.


Asunto(s)
Investigación Biomédica/organización & administración , Oncología Médica/organización & administración , Neoplasias/fisiopatología , Neoplasias/terapia , Antineoplásicos Inmunológicos/uso terapéutico , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epigénesis Genética , Genómica , Accesibilidad a los Servicios de Salud , Humanos , Invasividad Neoplásica/fisiopatología , Neoplasias/epidemiología , Neoplasias/genética , Células Madre Neoplásicas/fisiología
3.
Gac Med Mex ; 157(4): 454-462, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35133348

RESUMEN

Since its foundation, more than 75 years ago, the Mexican Institute of Social Security (IMSS) has carried out multidisciplinary research -biomedical, clinical and epidemiological- focused on understanding and solving the medical problems that afflict its beneficiaries (more than 50% of the Mexican population). Initially, research was the result of individual and isolated efforts. In the 1960s, a small number of researchers formed the first research groups. Currently, 240 full-time scientists work at five centers and 40 research units located in different states of Mexico. In addition, approximately 270 doctors carry out clinical research at different primary, secondary and tertiary care units. During these seven decades, IMSS scientists have made relevant contributions to medicine, which have not only helped increase knowledge on the etiopathogenesis of numerous diseases, but also their diagnosis, prognosis and treatment. This article presents an overview of medical research carried out at IMSS, based on a historical approach and a review of some of the most relevant contributions in different fields of research.


Desde su fundación, hace más de 75 años, el Instituto Mexicano del Seguro Social (IMSS) realiza investigación multidisciplinaria ­biomédica, clínica y epidemiológica­ enfocada a entender y resolver los problemas médicos que aquejan a sus derechohabientes (más de 50 % de la población mexicana). En un inicio, la investigación fue resultado de esfuerzos individuales y aislados. En la década de 1960, un número reducido de investigadores conformó los primeros grupos de investigación. Actualmente, 240 científicos de tiempo completo trabajan en cinco centros y 40 unidades de investigación ubicados en distintos estados de México. Además, aproximadamente 270 médicos efectúan investigación clínica en las distintas unidades de primer, segundo y tercer nivel de atención. Durante estas siete décadas, los científicos del IMSS han realizado aportaciones relevante para la medicina, las cuales no solo han ayudado a incrementar el conocimiento acerca de la etiopatogenia de numerosas enfermedades, sino también al diagnóstico, pronóstico y tratamiento de ellas. En este artículo se presenta un panorama general sobre la investigación médica que se desarrolla en el IMSS, a partir de un enfoque histórico y de la revisión de algunas de las contribuciones más relevantes en los distintos campos de la investigación.


Asunto(s)
Investigación Biomédica , Seguridad Social , Academias e Institutos , Humanos , Renta , México
4.
Blood Cells Mol Dis ; 85: 102485, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32836190

RESUMEN

In vitro growth of hematopoietic cells depends on the presence of hematopoietic cytokines. To date, it is unclear if these cells would be able to respond to non-hematopoietic cytokines. In the present study, we have explored this by culturing human hematopoietic cells in presence of neurogenic cytokines. Lineage-negative (Lin-) umbilical cord blood (UCB)-derived cells -enriched for hematopoietic stem and progenitor cells- were cultured in presence of different combinations of hematopoietic cytokines, neurotrophins, epidermal growth factor, fibroblast growth factor, and neurogenic culture media, in a 3-phase culture system. A proportion (1-22%) of Lin- UCB hematopoietic cells normally express neural markers and are capable of responding to neural cytokines. Neural cytokines did not have effects on hematopoietic cell proliferation; however, we observed generation of neural-like cells, assessed by morphology, and a significant increase in the proportion of cells expressing neural markers. Such neural-like cells, however, retained expression of hematopoietic markers. It seems that under our culture conditions, no actual transdifferentiation of hematopoietic cells into neural cells occurred; instead, the cells generated in culture seem to be hematopoietic cells that acquired neural features upon contact with neurogenic factors. The identity of UCB cells that acquired a neural phenotype is still unclear.


Asunto(s)
Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Neurogénesis , Técnicas de Cultivo de Célula , Células Cultivadas , Citocinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Factores de Crecimiento Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo
5.
J Cell Mol Med ; 22(10): 4899-4912, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30079458

RESUMEN

Tyrosine kinase inhibitors (TKI) have become a first-line treatment for chronic myeloid leuakemia (CML). TKIs efficiently target bulk CML cells; however, they are unable to eliminate the leukaemic stem cell (LSC) population that causes resistance and relapse in CML patients. In this study, we assessed the effects of parthenolide (PTL) and dimethyl amino parthenolide (DMAPT), two potent inhibitors of LSCs in acute myeloid leukaemia (AML), on CML bulk and CML primitive (CD34+ lin- ) cells. We found that both agents induced cell death in CML, while having little effect on the equivalent normal hematopoietic cells. PTL and DMAPT caused an increase in reactive oxygen species (ROS) levels and inhibited NF-κB activation. PTL and DMAPT inhibited cell proliferation and induced cell cycle arrest in G0 and G2 phases. Furthermore, we found cell cycle inhibition to correlate with down-regulation of cyclin D1 and cyclin A. In summary, our study shows that PTL and DMAPT have a strong inhibitory effect on CML cells. Given that cell cycle arrest was not dependent on ROS induction, we speculate that this effect could be a direct consequence of NF-κB inhibition and if this mechanism was to be evaded, PTL and DMAPT induced cell death would be potentiated.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Sesquiterpenos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/genética , Ciclina D1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , FN-kappa B/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Cytotherapy ; 20(11): 1345-1354, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30322708

RESUMEN

OBJECTIVE: Cell cycle plays a fundamental role in the physiology of hematopoietic stem and progenitor cells. In the present study we used a negative selection system to obtain an immature cell population-enriched for cord blood-derived CD34+ cells-and we determined its proliferation, expansion and differentiation patterns as a function of the cell cycle status. The effects of hydroxyurea (HU) were also assessed. RESULTS: As compared with cells in synthesis (S)/Gap2 (G2)/mitosis (M), cells in quiescent state (G0)/Gap1 (G1) showed a higher proliferation potential in vitro. At culture onset, G0, G1 and S/G2/M cells corresponded with 63%, 33% and 4%, respectively. Treatment with HU before culture resulted in an increase in the proportion of cells in G1 with a concomitant decrease in S/G2/M cells, without affecting the proportion of cells in G0. After 3 days of culture in the presence of recombinant cytokines, the vast majority of the cells (90%) were in G1, and by day 8, G0, G1 and S/G2/M cells corresponded with 18%, 67% and 15%, respectively. HU also induced an increase in colony-forming cell (CFC) frequency, in the proliferation and expansion capacities of cultured cells under myeloid conditions, and favored the development of the erythroid lineage. CONCLUSION: Our results show that the in vitro proliferation, expansion and differentiation potentials of immature hematopoietic cells are determined, at least in part, by their cell cycle status and that the cell cycle modifier HU significantly influences the growth of human hematopoietic cells. These results are of potential relevance for the development of ex vivo expansion protocols.


Asunto(s)
Antígenos CD34/metabolismo , Ciclo Celular/fisiología , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Hidroxiurea/farmacología , Células Sanguíneas/citología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Humanos , Cordón Umbilical/irrigación sanguínea
7.
Neurochem Res ; 42(2): 415-427, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27804011

RESUMEN

Neural progenitors (NP), found in fetal and adult brain, differentiate into neurons potentially able to be used in cell replacement therapies. This approach however, raises technical and ethical problems which limit their potential therapeutic use. Alternately, NPs can be obtained by transdifferentiation of non-neural somatic cells evading these difficulties. Human bone marrow mesenchymal stromal cells (MSCs) are suggested to transdifferentiate into NP-like cells, which however, have a low proliferation capacity. The present study demonstrates the requisite of cell adhesion for proliferation and survival of NP-like cells and re-evaluates some neuronal features after differentiation by standard procedures. Mature neuronal markers, though, were not detected by these procedures. A chemical differentiation approach was used in this study to convert MSCs-derived NP-like cells into neurons by using a cocktail of six molecules, CHIR99021, I-BET151, RepSox, DbcAMP, forskolin and Y-27632, defined after screening combinations of 22 small molecules. Direct transdifferentiation of MSCs into neuronal cells was obtained with the small molecule cocktail, without requiring the NP-like intermediate stage.


Asunto(s)
Proliferación Celular/fisiología , Transdiferenciación Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Adolescente , Adulto , Amidas/administración & dosificación , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Colforsina/administración & dosificación , Combinación de Medicamentos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Piridinas/administración & dosificación , Adulto Joven
8.
BMC Cancer ; 15: 722, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26475474

RESUMEN

BACKGROUND: The p38 MAPK is constitutively activated in B-NHL cell lines and regulates chemoresistance. Accordingly, we hypothesized that activated p38 MAPK may be associated with the in vivo unresponsiveness to chemotherapy in B-NHL patients. METHODS: Tissue microarrays generated from eighty untreated patients with Diffused Large B Cell Lymphoma (DLBCL) were examined by immunohistochemistry for the expression of p38 and phospho p38 (p-p38) MAPK. In addition, both Bcl-2 and NF-κB expressions were determined. Kaplan Meier analysis was assessed. RESULTS: Tumor tissues expressed p38 MAPK (82 %) and p-p38 MAPK (30 %). Both p38 and p-p38 MAPK expressions correlated with the high score performance status. A significant correlation was found between the expression p-p38 and poor response to CHOP. The five year median follow-up FFS was 81 % for p38(-) and 34 % for p38(+) and for OS was 83 % for p38(-) and 47 % for p38(+). The p-p38(+) tissues expressed Bcl-2 and 90 % of p-p38(-) where Bcl-2(-). The coexpression of p-p38 and Bcl-2 correlated with pool EFS and OS. There was no correlation between the expression of p-p38 and the expression of NF-κB. CONCLUSION: The findings revealed, for the first time, that a subset of patients with DLBCL and whose tumors expressed high p-p38 MAPK responded poorly to CHOP therapy and had poor EFS and OS. The expression of p38, p-p38, Bcl2 and the ABC subtype are significant risk factors both p38 and p-p38 expressions remain independent prognostic factors.


Asunto(s)
Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Doxorrubicina/administración & dosificación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Estimación de Kaplan-Meier , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad , FN-kappa B/genética , Prednisona/administración & dosificación , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Análisis de Matrices Tisulares , Vincristina/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/genética
9.
Transfusion ; 53(3): 668-78, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22845003

RESUMEN

BACKGROUND: Ex vivo expansion of hematopoietic stem and progenitor cells has become a priority in the experimental hematology arena. In this study we have obtained different hematopoietic cell populations from umbilical cord blood and simultaneously assessed their proliferation and expansion kinetics. Our main goal was to determine which one of these cell populations would be more suitable for clinical-grade ex vivo expansion. STUDY DESIGN AND METHODS: By using immunomagnetic-negative selection and cell sorting, five cell populations were obtained: unseparated mononuclear cells (MNCs; I); two lineage-negative cell populations, one enriched for CD34+ CD38+ cells (II) and the other enriched for CD34+ CD38- cells (III); and two CD34+ cell fractions purified by fluorescence-activated cell sorting, one containing CD34+ CD38+ cells (IV) and the other containing CD34+ CD38- cells (V). The kinetics of such populations were analyzed in both relative and absolute terms. RESULTS: No expansion was observed in Population I; in contrast, significant increments in the numbers of both progenitor and stem cells were observed in cultures of Populations II to V. Population V (reaching 12,800-fold increase in total cells; 1280-fold increase in CD34+ cells; 490-fold increase in colony-forming cells; and 12-fold increase in long-term culture-initiating cells) showed the highest proliferation and expansion potentials. CONCLUSION: Our study suggests that the cell fraction containing greater than 98% CD34+ CD38- cells would be the ideal one for large-scale ex vivo expansion; however, based on our data, it seems that, except for MNCs, all other cell populations could also be used as input cell fractions.


Asunto(s)
Técnicas de Cultivo de Célula , Proliferación Celular , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Antígenos CD34/metabolismo , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Separación Celular , Células Cultivadas , Conducta de Elección , Ensayo de Unidades Formadoras de Colonias/métodos , Sangre Fetal/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Cultivo Primario de Células/métodos , Factores de Tiempo
10.
Clin Dev Immunol ; 2013: 349067, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24198842

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most frequent malignancy of childhood. Substantial progress on understanding the cell hierarchy within ALL bone marrow (BM) has been recorded in the last few years, suggesting that both primitive cell fractions and committed lymphoid blasts with immature stem cell-like properties contain leukemia-initiating cells. Nevertheless, the biology of the early progenitors that initiate the lymphoid program remains elusive. The aim of the present study was to investigate the ability of lymphoid progenitors from B-cell precursor ALL BM to proliferate and undergo multilineage differentiation. By phenotype analyses, in vitro proliferation assays, and controlled culture systems, the lymphoid differentiation potentials were evaluated in BM primitive populations from B-cell precursor ALL pediatric patients. When compared to their normal counterparts, functional stem and progenitor cell contents were substantially reduced in ALL BM. Moreover, neither B nor NK or dendritic lymphoid-cell populations developed recurrently from highly purified ALL-lymphoid progenitors, and their proliferation and cell cycle status revealed limited proliferative capacity. Interestingly, a number of quiescence-associated transcription factors were elevated, including the transcriptional repressor Gfi-1, which was highly expressed in primitive CD34⁺ cells. Together, our findings reveal major functional defects in the primitive hematopoietic component of ALL BM. A possible contribution of high levels of Gfi-1 expression in the regulation of the stem/progenitor cell biology is suggested.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Leucémica de la Expresión Génica , Células Progenitoras Linfoides/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción/genética , Adolescente , Apoptosis , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Niño , Preescolar , Femenino , Humanos , Lactante , Células Progenitoras Linfoides/patología , Masculino , Fenotipo
11.
Gac Med Mex ; 149(6): 646-54, 2013.
Artículo en Español | MEDLINE | ID: mdl-24276188

RESUMEN

Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia associated with the t(9,22)(q34:q11) reciprocal translocation, also known as Philadelphia chromosome (Ph). As a result of such abnormality, a chimeric gene (bcr-abl) is produced that is translated into a chimeric protein (BCR-ABL), a constitutively activated tyrosine kinase. Major cell dysfunctions result from this abnormal kinase activity, including increased proliferation and reduced apoptosis. Based on the structure of BCR-ABL, several molecules have been designed that inhibit its kinase activity. Five such molecules have already been brought into the clinic for the treatment of Ph+ CML patients. Good results have been obtained in terms of patients' remission rates and quality of life. Some major problems, however, have been observed. Firstly, a significant proportion of patients develop resistance to the drugs; secondly, it is clear that such drugs affect most of the leukemic cells, but do not eliminate leukemia stem cells. Thus, important CML-related challenges remain to be solved in the near future.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Benzamidas/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Mesilato de Imatinib , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico
12.
Stem Cells Dev ; 32(7-8): 163-169, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36727603

RESUMEN

Cellular therapy (CT) can be defined as the transference into a person of healthy cells to correct defective functions. Yesterday (1950-2010), CT consisted mostly of hematopoietic transplants for the treatment of a variety of hematological disorders. Interestingly, during that period of time other cell types with therapeutic potential-including certain lymphoid populations and other nonhematopoietic cells-were discovered and characterized; thus, CT became a promising discipline for the treatment of a broader diversity of diseases. Today (2011-2023), CT has significantly grownup through preclinical studies and clinical trials, and it is currently progressing toward its consolidation as one of the pillars of medicine in the 21st century. Indeed, different types of stem cells (e.g., hematopoietic, mesenchymal, neural, and pluripotent), as well as different lymphoid and myeloid cell populations (e.g., TILs, CAR-Ts, CAR-NKs, and DUOC-01) are being used in clinical settings or are being tested in clinical trials. For the past decade, several CT modalities have been developed, and today, many of them are being used in the clinic. Tomorrow (2024-2040), already established CT modalities will surely be improved and applied more frequently, and novel therapies (that will include cell types such as iPSCs) will enter and expand within the clinical ground. It is noteworthy, however, that despite significant advancements and achievements, problems still need to be solved and obstacles need to be overcome. Technical, ethical, and economic issues persist and they need to be addressed. Undoubtedly, exciting times of challenges and opportunities are coming ahead in the CT arena.


Asunto(s)
Enfermedades Hematológicas , Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/trasplante
13.
J Leukoc Biol ; 114(5): 404-420, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37386890

RESUMEN

Blood cell formation (hematopoiesis) takes place mainly in the bone marrow, within the hematopoietic microenvironment, composed of a number of different cell types and their molecular products that together shape spatially organized and highly specialized microstructures called hematopoietic niches. From the earliest developmental stages and throughout the myeloid and lymphoid lineage differentiation pathways, hematopoietic niches play a crucial role in the preservation of cellular integrity and the regulation of proliferation and differentiation rates. Current evidence suggests that each blood cell lineage develops under specific, discrete niches that support committed progenitor and precursor cells and potentially cooperate with transcriptional programs determining the gradual lineage commitment and specification. This review aims to discuss recent advances on the cellular identity and structural organization of lymphoid, granulocytic, monocytic, megakaryocytic, and erythroid niches throughout the hematopoietic microenvironment and the mechanisms by which they interconnect and regulate viability, maintenance, maturation, and function of the developing blood cells.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Linaje de la Célula , Médula Ósea/metabolismo , Diferenciación Celular , Hematopoyesis , Células de la Médula Ósea
14.
Cells ; 12(7)2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37048119

RESUMEN

Macrophages with the M2 phenotype promote tumor development through the immunosuppression of antitumor immunity. We previously demonstrated the presence of mesenchymal stem/stromal cells (MSCs) in cervical cancer (CeCa-MSCs), suggesting an immune protective capacity in tumors, but to date, their effect in modulating macrophage polarization remains unknown. In this study, we compared the capacities of MSCs from normal cervix (NCx) and CeCa to promote M2 macrophage polarization in a coculture system. Our results demonstrated that CeCa-MSCs, in contrast to NCx-MSCs, significantly decreased M1 macrophage cell surface marker expression (HLA-DR, CD80, CD86) and increased M2 macrophage expression (CD14, CD163, CD206, Arg1) in cytokine-induced CD14+ monocytes toward M1- or M2-polarized macrophages. Interestingly, compared with NCx-MSCs, in M2 macrophages generated from CeCa-MSC cocultures, we observed an increase in the percentage of phagocytic cells, in the intracellular production of IL-10 and IDO, the capacity to decrease T cell proliferation and for the generation of CD4+CD25+FoxP3+ Tregs. Importantly, this capacity to promote M2 macrophage polarization was correlated with the intracellular expression of macrophage colony-stimulating factor (M-CSF) and upregulation of IL-10 in CeCa-MSCs. Furthermore, the presence of M2 macrophages was correlated with the increased production of IL-10 and IL-1RA anti-inflammatory molecules. Our in vitro results indicate that CeCa-MSCs, in contrast to NCx-MSCs, display an increased M2-macrophage polarization potential and suggest a role of CeCa-MSCs in antitumor immunity.


Asunto(s)
Interleucina-10 , Neoplasias del Cuello Uterino , Humanos , Femenino , Interleucina-10/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Macrófagos/metabolismo , Citocinas/metabolismo , Células del Estroma/metabolismo
15.
J Leukoc Biol ; 114(6): 672-683, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-37820030

RESUMEN

Neutrophils infiltrate several types of cancer; however, whether their presence is associated with disease progression remains controversial. Here, we show that colon tumors overexpress neutrophil chemoattractants compared to healthy tissues, leading to their recruitment to the invasive margin and the central part of colon tumors. Of note, tumor-associated neutrophils expressing tumor necrosis factor α, which usually represents an antitumoral phenotype, were predominantly located in the invasive margin. Tumor-associated neutrophils from the invasive margin displayed an antitumoral phenotype with higher ICAM-1 and CD95 expression than neutrophils from healthy adjacent tissues. A higher neutrophil/lymphocyte ratio was found at later stages compared to the early phases of colon cancer. A neutrophil/lymphocyte ratio ≤3.5 predicted tumor samples had significantly more neutrophils at the invasive margin and the central part. Moreover, tumor-associated neutrophils at the invasive margin of early-stage tumors showed higher ICAM-1 and CD95 expression. Coculture of colon cancer cell lines with primary neutrophils induced ICAM-1 and CD95 expression, confirming our in situ findings. Thus, our data demonstrate that tumor-associated neutrophils with an antitumoral phenotype characterized by high ICAM-1 and CD95 expression infiltrate the invasive margin of early-stage colon tumors, suggesting that these cells can combat the disease at its early courses. The presence of tumor-associated neutrophils with antitumoral phenotype could help predict outcomes of patients with colon cancer.


Asunto(s)
Neoplasias del Colon , Neutrófilos , Humanos , Neutrófilos/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Neoplasias del Colon/patología , Fenotipo
16.
Arch Med Res ; 53(8): 747-752, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36460549

RESUMEN

During the last five decades different therapies have been developed for the treatment of cancer, and as a result, patients can now live longer and better lives. Among such therapies, hematopoietic cell transplantation and immunotherapy have played key roles. In this short article, we present our particular point of view on the development of these two cellular therapies. We have focused on a historical perspective emphasizing the work of some of the Nobel Prize winners whose studies constituted cornerstones in our knowledge of the biology of cancer and in our fight against this devastating disease.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias , Humanos , Historia del Siglo XX , Premio Nobel , Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia , Neoplasias/terapia
17.
Arch Med Res ; 53(8): 770-784, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36462951

RESUMEN

It is well recognized that most cancers derive and progress from transformation and clonal expansion of a single cell that possesses stem cell properties, i.e., self-renewal and multilineage differentiation capacities. Such cancer stem cells (CSCs) are usually present at very low frequencies and possess properties that make them key players in tumor development. Indeed, besides having the ability to initiate tumor growth, CSCs drive tumor progression and metastatic dissemination, are resistant to most cancer drugs, and are responsible for cancer relapse. All of these features make CSCs attractive targets for the development of more effective oncologic treatments. In the present review article, we have summarized recent advances in the biology of CSCs, including their identification through their immunophenotype, and their physiology, both in vivo and in vitro. We have also analyzed some molecular markers that might become targets for developing new therapies aiming at hampering CSCs regeneration and cancer relapse.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Células Madre Neoplásicas , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Diferenciación Celular
18.
Hematology ; 27(1): 476-487, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35413231

RESUMEN

OBJECTIVE: The interplay between intrinsic and extrinsic elements involved in the physiology of hematopoietic cells is not completely understood. In the present study, we analyzed the transcriptional profiles of human cord blood-derived hematopoietic stem cells (HSCs), as well as myeloid (MPCs) and erythroid (EPCs) progenitors, and assessed their proliferation and expansion kinetics in vitro. METHODS: All cell populations were obtained by cell-sorting, and were cultured in liquid cultures supplemented with different cytokine combinations. Their gene expression profiles were determined by RNA microarrays right after cell-sorting, before culture. RESULTS: HSCs showed the highest proliferation and expansion capacities in culture, and were found to be more closely related, in transcriptional terms, to MPCs than to EPCs. This correlated with the fact that after 30 days, only cultures initiated with HSCs and MPCs were sustained. Expression of cell cycle and cell division-related genes was enriched in EPCs. Such cells showed significantly higher proliferation than MPCs, however, their expansion potential was reduced, so that cultures initiated with EPCs declined after 15 days and became exhausted by day 30. Proliferation and expansion of HSCs and EPCs were higher in the presence of a cytokine combination that favors erythropoiesis, whereas the growth of MPCs was higher under a cytokine combination that favors myelopoiesis. CONCLUSION: This study shows a correlation between the transcriptional profiles of HSCs, MPCs, and EPCs, and their respective in vitro growth under particular culture conditions. These results may be relevant in the development of ex vivo systems for the expansion of hematopoietic cells for clinical application.


Asunto(s)
Citocinas , Células Madre Hematopoyéticas , Antígenos CD34/metabolismo , Proliferación Celular , Células Cultivadas , Citocinas/genética , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Transcriptoma
19.
J Leukoc Biol ; 112(1): 31-45, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35674096

RESUMEN

Leukemogenesis is proposed to result from the continuous interplay between inducive bone marrow (BM) microenvironments and malignant precursor cells. Recent findings point toward an abnormal production of proinflammatory mediators within the BM from acute lymphoblastic leukemia (ALL) patients, although the mechanism underlying this phenomenon is uncertain. Here, we have identified 3 miRNAs, miR-146a-5p, miR-181b-5p, and miR-199b-3p, as potential candidates for TLR8 ligation, which are overexpressed in ALL and show agonist functional binding. When purified from ALL exosomes, they demonstrated their capacity of inducing cytokine production by both, hematopoietic and stromal BM cells. Of note, the exposure of BM cells from ALL patients to the proinflammatory milieu resulting from these miRNAs agonist activity revealed the proliferation of normal progenitors, while poor effects were recorded in the leukemic counterpart. The unconventional roles of the tumor-secreted miRNAs as TLR8 agonist ligands may provide a novel mechanism contributing a tumor-microenvironment feedback loop by switching on proinflammatory pathways that further activate normal hematopoietic precursors and support ALL progression. Secreted B-ALL TLR8-agonist miRNAs are involved in the promotion of proinflammatory microenvironments that target normal hematopoietic cells. B-lineage ALL cells secrete exosomes containing miRNAs endowed with the ability of functionally binding TLR8 in hematopoietic and BM mesenchymal stromal cells. Upon TLR8 signaling, the activation of the NF-kB pathway induces secretion of proinflammatory cytokines that, in turn, promotes cell proliferation in early hematopoietic cell populations, driving a tumor-microenvironment-hematopoietic activation feedback loop that may reduce the normal hematopoietic stem and progenitor cell compartment and facilitate cancer progression.


Asunto(s)
MicroARNs , Leucemia-Linfoma Linfoblástico de Células Precursoras , Médula Ósea/patología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptor Toll-Like 8/metabolismo , Microambiente Tumoral
20.
Eur J Histochem ; 66(1)2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35174683

RESUMEN

Desmoplastic stroma (DS) and the epithelial-to-mesenchymal transition (EMT) play a key role in pancreatic ductal adenocarcinoma (PDAC) progression. To date, however, the combined expression of DS and EMT markers, and their association with variations in survival within each clinical stage and degree of tumor differentiation is unknown. The purpose of this study was to investigate the association between expression of DS and EMT markers and survival variability in patients diagnosed with PDAC. We examined the expression levels of DS markers alpha smooth muscle actin (α-SMA), fibronectin, and vimentin, and the EMT markers epithelial cell adhesion molecule (EPCAM), pan-cytokeratin, and vimentin, by immunohistochemistry using a tissue microarray of a retrospective cohort of 25 patients with PDAC. The results were examined for association with survival by clinical stage and by degree of tumor differentiation. High DS markers expression -α-SMA, fibronectin, and vimentin- was associated with decreased survival at intermediate and advanced clinical stages (p=0.006-0.03), as well as with both poorly and moderately differentiated tumor grades (p=0.01-0.02). Interestingly, the same pattern was observed for EMT markers, i.e., EPCAM, pan-cytokeratin, and vimentin (p=0.00008-0.03). High expression of DS and EMT markers within each clinical stage and degree of tumor differentiation was associated with lower PDAC survival. Evaluation of these markers may have a prognostic impact on survival time variation in patients with PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Transición Epitelial-Mesenquimal/fisiología , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA