Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Blood Cells Mol Dis ; 110: 102895, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39303397

RESUMEN

Small molecules UM171 and SR1 have already been taken into clinically-oriented protocols for the ex vivo expansion of hematopoietic stem (HSCs) and progenitor (HPCs) cells. In order to gain further insight into their biology, in the present study we have assessed their effects, both individually and in combination, on the in vitro long-term proliferation and expansion of HSCs and HPCs contained within three different cord blood-derived cell populations: MNCs (CD34+ cells = 0.8 %), LIN- cells (CD34+ cells = 41 %), and CD34+ cells (CD34+ cells >98 %). Our results show that when added to cultures in the absence of recombinant stimulatory cytokines, neither molecule had any effect. In contrast, when added in the presence of hematopoietic cytokines, UM171 and SR1 had significant stimulatory effects on cell proliferation and expansion in cultures of LIN- and CD34+ cells. No significant effects were observed in cultures of MNCs. The effects of both molecules were more pronounced in cultures with the highest proportion of CD34+ cells, and the greatest effects were observed when both molecules were added in combination. In the absence of small molecules, cell numbers reached a peak by days 25-30, and then declined; whereas in the presence of UM171 or/and SR1 cell numbers were sustained up to day 45 of culture. Our results indicate that besides CD34+ cells, LIN- cells could also be used as input cells in clinically-oriented expansion protocols, and that using both molecules simultaneously would be a better approach than using only one of them.

2.
Int J Mol Sci ; 25(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39273524

RESUMEN

Human dental tissue mesenchymal stem cells (DT-MSCs) constitute an attractive alternative to bone marrow-derived mesenchymal stem cells (BM-MSCs) for potential clinical applications because of their accessibility and anti-inflammatory capacity. We previously demonstrated that DT-MSCs from dental pulp (DP-MSCs), periodontal ligaments (PDL-MSCs), and gingival tissue (G-MSCs) show immunosuppressive effects similar to those of BM, but to date, the DT-MSC-mediated immunoregulation of T lymphocytes through the purinergic pathway remains unknown. In the present study, we compared DP-MSCs, PDL-MSCs, and G-MSCs in terms of CD26, CD39, and CD73 expression; their ability to generate adenosine (ADO) from ATP and AMP; and whether the concentrations of ADO that they generate induce an immunomodulatory effect on T lymphocytes. BM-MSCs were included as the gold standard. Our results show that DT-MSCs present similar characteristics among the different sources analyzed in terms of the properties evaluated; however, interestingly, they express more CD39 than BM-MSCs; therefore, they generate more ADO from ATP. In contrast to those produced by BM-MSCs, the concentrations of ADO produced by DT-MSCs from ATP inhibited the proliferation of CD3+ T cells and promoted the generation of CD4+CD25+FoxP3+CD39+CD73+ Tregs and Th17+CD39+ lymphocytes. Our data suggest that DT-MSCs utilize the adenosinergic pathway as an immunomodulatory mechanism and that this mechanism is more efficient than that of BM-MSCs.


Asunto(s)
5'-Nucleotidasa , Adenosina , Apirasa , Pulpa Dental , Células Madre Mesenquimatosas , Ligamento Periodontal , Linfocitos T , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Humanos , Adenosina/metabolismo , Pulpa Dental/citología , Pulpa Dental/inmunología , Pulpa Dental/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , 5'-Nucleotidasa/metabolismo , Apirasa/metabolismo , Ligamento Periodontal/citología , Ligamento Periodontal/metabolismo , Adenosina Trifosfato/metabolismo , Células Cultivadas , Encía/citología , Encía/metabolismo , Encía/inmunología , Antígenos CD/metabolismo , Inmunomodulación , Diferenciación Celular , Proliferación Celular , Dipeptidil Peptidasa 4/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Proteínas Ligadas a GPI
3.
Arch Med Res ; 55(6): 103042, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39003965

RESUMEN

Umbilical cord blood (UCB) is a rich source of hematopoietic stem and progenitor cells that are biologically superior to their adult counterparts. UCB cells can be stored for several years without compromising their numbers or function. Today, public and private UCB banks have been established in several countries around the world. After 35 years since the first UCB transplant (UCBT), more than 50,000 UCBTs have been performed worldwide. In pediatric patients, UCBT is comparable to or superior to bone marrow transplantation. In adult patients, UCB can be an alternative source of hematopoietic cells when an HLA-matched unrelated adult donor is not available and when a transplant is urgently needed. Delayed engraftment (due to reduced absolute numbers of hematopoietic cells) and higher costs have led many medical institutions not to consider UCB as a first-line cell source for hematopoietic transplants. As a result, the use of UCB as a source of hematopoietic stem and progenitor cells for transplantation has declined over the past decade. Several approaches are being investigated to make UCBTs more efficient, including improving the homing capabilities of primitive UCB cells and increasing the number of hematopoietic cells to be infused. Several of these approaches have already been applied in the clinic with promising results. UCB also contains immune effector cells, including monocytes and various lymphocyte subsets, which, together with stem and progenitor cells, are excellent candidates for the development of cellular therapies for hematological and non-hematological diseases.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos
4.
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
6.
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
7.
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
8.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
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
16.
STAR Protoc ; 2(4): 100846, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34622219

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow and supply blood cells. Efficient methods for isolation of HSPCs are required. Here, we present protocols for the isolation of human and murine HSPCs using manual and FACS-assisted techniques. Isolated HSPCs can be used for downstream applications, including colony forming unit assays and DNA damage and repair assays. For complete details on the use and execution of this protocol, please refer to Rodríguez et al. (2021a) and (2021b).


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Ensayo de Unidades Formadoras de Colonias , Daño del ADN/genética , Reparación del ADN , Humanos , Ratones
17.
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
18.
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
19.
Ann Med ; 53(1): 197-207, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33345622

RESUMEN

BACKGROUND: COVID-19 counts 46 million people infected and killed more than 1.2 million. Hypoxaemia is one of the main clinical manifestations, especially in severe cases. HIF1α is a master transcription factor involved in the cellular response to oxygen levels. The immunopathogenesis of this severe form of COVID-19 is poorly understood. METHODS: We performed scRNAseq from leukocytes from five critically ill COVID-19 patients and characterized the expression of hypoxia-inducible factor1α and its transcriptionally regulated genes. Also performed metanalysis from the publicly available RNAseq data from COVID-19 bronchoalveolar cells. RESULTS: Critically-ill COVID-19 patients show a shift towards an immature myeloid profile in peripheral blood cells, including band neutrophils, immature monocytes, metamyelocytes, monocyte-macrophages, monocytoid precursors, and promyelocytes-myelocytes, together with mature monocytes and segmented neutrophils. May be the result of a physiological response known as emergency myelopoiesis. These cellular subsets and bronchoalveolar cells express HIF1α and their transcriptional targets related to inflammation (CXCL8, CXCR1, CXCR2, and CXCR4); virus sensing, (TLR2 and TLR4); and metabolism (SLC2A3, PFKFB3, PGK1, GAPDH and SOD2). CONCLUSIONS: The up-regulation and participation of HIF1α in events such as inflammation, immunometabolism, and TLR make it a potential molecular marker for COVID-19 severity and, interestingly, could represent a potential target for molecular therapy. Key messages Critically ill COVID-19 patients show emergency myelopoiesis. HIF1α and its transcriptionally regulated genes are expressed in immature myeloid cells which could serve as molecular targets. HIF1α and its transcriptionally regulated genes is also expressed in lung cells from critically ill COVID-19 patients which may partially explain the hypoxia related events.


Asunto(s)
COVID-19/genética , Enfermedad Crítica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Mieloides/metabolismo , Análisis de Secuencia de ARN/métodos , Femenino , Humanos , Masculino , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
20.
Arch Med Res ; 52(3): 311-323, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33248817

RESUMEN

BACKGROUND: SARS-CoV-2, the etiological agent causing COVID-19, has infected more than 27 million people with over 894000 deaths worldwide since its emergence in December 2019. Factors for severe diseases, such as diabetes, hypertension, and obesity have been identified however, the precise pathogenesis is poorly understood. To understand its pathophysiology and to develop effective therapeutic strategies, it is essential to define the prevailing immune cellular subsets. METHODS: We performed whole circulating immune cells scRNAseq from five critically ill COVID-19 patients, trajectory and gene ontology analysis. RESULTS: Immature myeloid populations, such as promyelocytes-myelocytes, metamyelocytes, band neutrophils, monocytoid precursors, and activated monocytes predominated. The trajectory with pseudotime analysis supported the finding of immature cell states. While the gene ontology showed myeloid cell activation in immune response, DNA and RNA processing, defense response to the virus, and response to type 1 interferon. Lymphoid lineage was scarce. Expression of genes such as C/EBPß, IRF1and FOSL2 potentially suggests the induction of trained immunity. CONCLUSIONS: Our results uncover transcriptomic profiles related to immature myeloid lineages and suggest the potential induction of trained immunity.


Asunto(s)
COVID-19/sangre , Células Mieloides/patología , COVID-19/patología , COVID-19/virología , Enfermedad Crítica , Humanos , SARS-CoV-2/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...