Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Microb Pathog ; 179: 106109, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37040845

RESUMEN

Human Cytomegalovirus (HCMV) infection is associated with bad obstetric history (BOH) and adverse pregnancy outcomes (APO). Here, we characterized antiviral humoral profiles, systemic and virus specific cellular immune responses concurrently in pregnant women (n = 67) with complications including BOH and associated these signatures with pregnancy outcomes. Infection status was determined using nested blood PCR, seropositivity and IgG avidity by ELISA. Systemic and HCMV specific (pp65) cellular immune responses were evaluated by flow cytometry. Seropositivity was determined for other TORCH pathogens (n = 33) on samples with recorded pregnancy outcomes. This approach was more sensitive in detecting HCMV infection. Blood PCR positive participants, irrespective of their IgG avidity status, had higher cytotoxic potential in circulating CD8+ T cells (p < 0.05) suggesting that infection associated cellular dysfunction was uncoupled with avidity maturation of antiviral humoral responses. Also, impaired anamnestic degranulation of HCMV-pp65-specific T cells compared to HCMV blood PCR negative participants (p < 0.05) was observed. APO correlated with HCMV blood PCR positivity but not serostatus (p = 0.0039). Most HCMV IgM positive participants (5/6) were HCMV blood PCR positive with APO. None were found to be IgM positive for other TORCH pathogens. Multiple TORCH seropositivity however was significantly enriched in the APO group (p = 0.024). Generation of HCMV specific high avidity IgG antibodies had no bearing on APO (p = 0.9999). Our study highlights the utility of an integrated screening approach for antenatal HCMV infection in the context of BOH, where infection is associated with systemic and virus specific cellular immune dysfunction as well as APO.


Asunto(s)
Infecciones por Citomegalovirus , Complicaciones Infecciosas del Embarazo , Embarazo , Humanos , Femenino , Resultado del Embarazo , Mujeres Embarazadas , Infecciones por Citomegalovirus/diagnóstico , Linfocitos T CD8-positivos , Monitorización Inmunológica , Citomegalovirus , Anticuerpos Antivirales , Inmunoglobulina G , Inmunoglobulina M
2.
Environ Toxicol ; 29(1): 84-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21987380

RESUMEN

Long-term exposure of agriculturally used organochloride and organophosphate pesticides have been shown to cause long-lasting hematotoxicity and increased incidence of aplastic anemia in humans. The mechanisms involved in pesticide induced hematotoxicity and the features of toxicity that may play a major role in bone marrow suppression are not known. The aim of the present study was to investigate the hematological consequences of pesticide exposure in swiss albino mice exposed to aqueous mixture of common agriculturally used pesticides for 6 h/day, 5 days/week for 13 weeks. After the end of last exposure, without a recovery period, the strong hematotoxic effect of pesticide was assessed in mice with long-term bone marrow explant culture (LTBMC-Ex) system and cell colony forming assays. Bone marrow explant culture from the pesticide exposed group of mice failed to generate a supportive stromal matrix and did not produce adequate number of hematopoietic cells and found to contain largely the adipogenic precursors. The decreased cell colony numbers in the pesticide exposed group indicated defective maturational and functional status of different marrow cell lineages. As a whole, exposure of mice to the mixture of pesticides reduced the total number of bone marrow cells (granulocytes are the major targets of pesticide toxicity), hematopoietic, and non-hematopoietic progenitor cells and most of the hematological parameters. Replication of primitive stem/progenitor cells in the marrow was decreased following pesticide exposure with G0/G1-phase arrest of most of the cells. The progenitor cells showed decreased percentage of cells in S/G2/M-phase. The increased apoptosis profile of the marrow progenitors (Increased CD95 expression) and primitive stem cells (High Annexin-V positivity on Sca1+ cells) with an elevated intracellular cleaved caspase-3 level on the Sca1+ bone marrow cells provided the base necessary for explaining the deranged bone marrow microenvironmental structure which was evident from scanning electron micrographs. These results clearly indicate a strong, long lasting toxic effect of pesticides on the bone marrow microenvironment and different microenvironmental components which ultimately leads to the formation of a degenerative disease like aplastic anemia.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Insecticidas/toxicidad , Células Madre Mesenquimatosas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Caspasa 3/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Ratones
3.
Biochem Cell Biol ; 91(3): 165-75, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23668789

RESUMEN

Good vision requires a healthy cornea, and a healthy cornea needs healthy stem cells. Limbal epithelial stem cells (LESCs) are a traditional source of corneal epithelial cells and are recruited for the continuous production of epithelium without seizing throughout an animal's life, which maintains corneal transparency. Like the maintenance of other adult somatic stem cells, the maintenance of LESCs depends on the specific microenvironmental niche in which they reside. The purpose of this study was to determine the microenvironmental damage associated with LESCs fate due to ultraviolet (UV)-B exposure in a mouse model. Structural alteration and deregulation of the stem cell and its neighboring niche components were observed by using clinical, morphological, explant culture study, and flowcytometric analysis, which demonstrated that the limbal microenvironment plays an important role in cornea-related disease development. In UV-exposed mice, overexpression of vascular endothelial growth factor receptor 2 indicated neovascularization, decreased CD38 expression signified the alteration of limbal epithelial superficial cells, and the loss of limbal stem cell marker p63 indicated limbal stem cell deficiency in the limbal vicinity. We concluded that LESC deficiency diseases (LESCDDs) are associated with pathophysiological changes in the LESC niche, with some inhibitory interception such as UV-B irradiation, which results in corneal defects.


Asunto(s)
Epitelio/efectos de la radiación , Células Madre/efectos de la radiación , Rayos Ultravioleta , Animales , Femenino , Masculino , Ratones , Microscopía Electrónica de Rastreo , Fenotipo
4.
Microbes Infect ; 25(7): 105165, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37247806

RESUMEN

The interplay of active HCMV infection with gut dysbiosis in the immunopathology of cholestasis in neonates and infants remains unexplored. In this study, we evaluated gut microbiome profiles and immune dysfunction in a cohort of HCMV infected cholestatic infants (IgM positive, N = 21; IgM negative, N = 25) compared to healthy infants, N = 10. HCMV infected IgM positive individuals exhibited increased clinical severity in terms of liver dysfunction, altered CD4+: CD8+ ratio, and elevated Granzyme B levels in cellular immune subsets. Gut microbiome analysis revealed distinct and differential diversity and composition within infected groups aligned with clinical severity reflected through the increased abundance of Gammaproteobacteria, reduced Bifidobacteria, and a unique signature mapping to the HCMV infected IgM negative group. Correlation analyses revealed associations between Bifidobacterium breve, Gammaproteobacteria, Firmicutes, Clostridia, Finegoldia magna, Veillonella dispar, and Granzyme B expressing immune cell subsets. Our study describes a novel gut microbiome-immune axis that may influence disease severity in cholestatic infants with active HCMV infection.


Asunto(s)
Colestasis , Infecciones por Citomegalovirus , Microbioma Gastrointestinal , Hepatopatías , Recién Nacido , Humanos , Lactante , Granzimas , Colestasis/microbiología , Inmunoglobulina M
5.
Chin J Cancer ; 29(12): 969-79, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21114916

RESUMEN

BACKGROUND AND OBJECTIVE: Leukemic microenvironment has a major role in the progression of leukemia. Leukemic cells can induce reversible changes in microenvironmental components, especially the stromal function which results in improved growth conditions for maintaining the malignant leukemic cells. This study aimed to investigate the survival advantage of leukemic cells over normal hematopoietic cells in stromal microenvironment in long term. METHODS: The mice were injected intraperitoneally with N-N' ethylnitrosourea (ENU) to induce leukemia; the mice received injection of normal saline were used as control. At 180 days after ENU induction, the mice were killed and the bone marrows were cultured for 19 days. Colony-forming assays were used to analyze the formation of various cell colonies. The expression of Sca-1, CD146, VEGFR2, CD95, pStat3, pStat5, and Bcl-xL in marrow cells were detected by flow cytometry. RESULTS: Long-term leukemic bone marrow culture showed abnormal elongated stromal fibroblasts with almost absence of normal hematopoietic cells. Adherent cell colonies were increased, but CFU-F and other hematopoietic cell colonies were significantly decreased in leukemia group (P<0.001). Primitive progenitor-specific Sca-1 receptor expression was decreased with subsequent increased expression of CD146 and VEGFR-2 in leukemic bone marrow cells. Decreased Fas antigen expression with increased intracellular pStat3, pStat5 and Bcl-xL proteins were observed in leukemic bone marrow cells. CONCLUSIONS: Stromal microenvironment shows altered morphology and decreased maturation in leukemia. Effective progenitor cells are decreased in leukemia with increased leukemia-specific cell population. Leukemic microenvironment plays a role in promoting and maintaining the leukemic cell proliferation and survivability in long term.


Asunto(s)
Células de la Médula Ósea/patología , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucemia/patología , Microambiente Tumoral/fisiología , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/metabolismo , Antígeno CD146/metabolismo , Recuento de Células , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Etilnitrosourea , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Células Progenitoras de Granulocitos y Macrófagos/patología , Granulocitos/metabolismo , Granulocitos/patología , Células Madre Hematopoyéticas/metabolismo , Leucemia/inducido químicamente , Leucemia/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Fenotipo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína bcl-X/metabolismo , Receptor fas/metabolismo
6.
Biol Trace Elem Res ; 181(2): 304-313, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28516389

RESUMEN

Exposure to arsenic on a regular basis, mainly through drinking water, agricultural pesticide, and sometimes therapeutic dose, results in various diseases of different tissues including the bone marrow hematopoietic system. Hematopoiesis is a dynamic process by which bone marrow (BM) hematopoietic stem/progenitor cells (HSPCs) generate a relatively constant pool of functionally mature blood cells by the support of microenvironmental components. The present study has been aimed to understand stem cell microenvironmental status during arsenic toxicity and the consequent reflection of dysregulation involving the hematopoietic machinery in experimental mice. Swiss albino mice were experimentally exposed to 10 µg arsenic trioxide/g body weight through oral gavage and 5 µg arsenic trioxide/g body weight intraperitoneally for a period of 30 days. Altered hemogram values in peripheral blood reflected the impaired hematopoiesis which was further validated by the reduced BM cellularity along with the deviated BM cell morphology as observed by scanning electron microscopy post arsenic exposure. The stromal cells were unable to establish a healthy matrix and the sustainability of hematopoietic progenitors was drastically affected in arsenic-exposed mouse groups, as observed in in vitro explant culture. The inability of stromal cells to establish supportive matrix was also explained by the decreased adherent colony formation in treated animals. Furthermore, the flow cytometric characterization of CXCR4+ and Sca-1+ CD44+ receptor expressions confirmed the dysregulation in the hematopoietic microenvironment. Thus, considering the importance of microenvironment in the maintenance of HSPC, it can be concluded that arsenic toxicity causes microenvironmental damage, leading to niche derangement and impaired hematopoiesis.


Asunto(s)
Arsénico/toxicidad , Médula Ósea/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Nicho de Células Madre/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Animales , Arsénico/administración & dosificación , Médula Ósea/metabolismo , Ratones , Receptores CXCR4/metabolismo
7.
Exp Toxicol Pathol ; 65(3): 287-95, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22005501

RESUMEN

Long term inhalation of toxic pesticides used for the domestic and industrial purposes have been shown to cause moderate to severe hematotoxicity and increased incidence of several marrow degenerative diseases, specifically hypoplastic bone marrow failure condition in humans. The progression of pesticide induced hematotoxicity and the exact underlying mechanisms of toxicity that play major role in limiting normal hematopoiesis are not quite well explained. In this present study, we have developed an animal model of hypoplastic bone marrow failure following pesticide exposure to show the deleterious effects of toxic pesticides on mouse hematopoietic system. Here we have presented the results of studying long-term marrow explant culture, IL-2, IL-3 and IL-5 receptors expression profile, fibroblast colony forming unit (CFU-F), hematopoietic progenitor cell colony formation and caspase-3 expression by the bone marrow cells. We have also identified the expression levels of several extracellular apoptosis markers (CD95/Fas) and intracellular apoptosis inducer proteins (pASK1, pJNK, caspase-3 and cleaved caspase-3) in the bone marrow cells of pesticide exposed mice. The long-term marrow explant culture demonstrated the impairment in proliferation of the stromal cells/stromal fibroblasts in culture. Decreased IL-2, IL-3 and IL-5 receptors expression profile essentially hinted at the suppressed cytokine activity in the pesticide exposed marrow. CFU-F analysis showed the defect in functional maturation of the stromal fibroblasts. The decreased hematopoietic progenitor cell colony formation indicated the toxicity induced inhibition of cellular proliferation and functional maturation of hematopoietic stem/progenitor cells in pesticide exposed marrow. We have detected a sharp increase in the expression levels of both the extracellular Fas-antigen and intracellular apoptosis inducer proteins in the bone marrow cells of pesticide exposed mice that explained well, the apoptosis pathway involved following marrow toxicity. The decreased proliferation and functional maturation of marrow stromal cells and hematopoietic progenitors with subsequent increase in marrow cellular apoptosis following pesticide toxicity provided the base necessary for explaining the increased incidence of hypoplastic bone marrow failure in humans exposed to moderate to high concentrations of pesticides.


Asunto(s)
Apoptosis/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Cloropirifos/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Insecticidas/toxicidad , Piretrinas/toxicidad , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Técnicas de Cultivo de Célula , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Ratones , Receptores de Interleucina-2/biosíntesis , Receptores de Interleucina-3/biosíntesis , Receptores de Interleucina-5/biosíntesis
8.
Arh Hig Rada Toksikol ; 63(3): 271-82, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23152377

RESUMEN

The mechanistic interplay between pesticide exposure and development of marrow aplasia is not yet well established but there are indices that chronic pesticide exposure in some instances causes marrow aplasia like haematopoietic degenerative condition in human beings. Canonical Hedgehog (Hh) signalling has multiple roles in a wide range of developmental processes, including haematopoiesis. The present study was designed to explore the status of four important components of the canonical Hedgehog signalling cascade, the Sonic Hedgehog (Shh), Ptch1, Smo, and Gli1, in a mouse model of chronic pesticide-induced bone marrow aplasia. We used 5 % aqueous mixture of pesticides (chlorpyriphos, prophenophos, cypermethrin, alpha-methrin, and hexaconazole) for inhalation and dermal exposure of 6 hours per day and 5 days a week up to 90 days. Murine bone marrow aplasia related to chronic pesticide treatment was confirmed primarily by haemogram, bone marrow cellularity, short term bone marrow explant culture for cellular kinetics, bone marrow smear, and fl ow cytometric Lin-Sca-1+C-kit+ extracellular receptor expression pattern. Later, components of hedgehog signalling were analysed in the bone marrow of both control and pesticide-treated aplastic groups of animals. The results depicted pancytopenic feature of peripheral blood, developmental anomaly of neutrophils, depression of primitive stem and progenitor population along with Shh, Ptch1, Smo and Gli1 expression in aplasia group. This investigation suggests that pesticide-induced downregulation of two critically important proteins--Ptch1 and Gli1--inside the haematopoietic stem and progenitor cell population impairs haematopoietic homeostasis and regeneration mechanism in vivo concurrent with bone marrow aplasia.


Asunto(s)
Enfermedades de la Médula Ósea/inducido químicamente , Enfermedades de la Médula Ósea/genética , Proteínas Hedgehog/genética , Insecticidas/toxicidad , Factores de Transcripción de Tipo Kruppel/genética , Receptores de Superficie Celular/genética , Transducción de Señal/genética , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Ratones , Receptores Patched , Receptor Patched-1 , Proteína con Dedos de Zinc GLI1
9.
J Stem Cells ; 5(3): 117-27, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-22314828

RESUMEN

Apoptosis, proliferation and differentiation are balanced molecular processes which may alter their pattern during environmental insults. Arsenic is an environmental pollutant, ranks 20(th) in abundance in the earth crust, 14(th) in sea water and 12(th) in the human body. Millions of people worldwide are chronically exposed to arsenic often due to naturally occurring arsenic in ground water. Hematopoietic stem cells within the bone marrow are the source of all haematopoietic cell lineages and are essential for tissue development throughout the life. In this experimental study, we have evaluated the impact of arsenic, on blood and blood forming cells by the changes in their cellular morphology, immune functional capacity, alteration of bone marrow CD34 positive stem/progenitors and changes in the phenotype of Sca-1, c-Kit dual positive primitive stem cell population. The study revealed that arsenic has a significant effect on bone marrow and hematopoietic stem cells, their immune capacity and upregulation of death process, all indicative of impairment in differentiation suggesting presence of deregulation in their precursors by arsenic toxicity.


Asunto(s)
Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Óxidos/toxicidad , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Apoptosis/fisiología , Trióxido de Arsénico , Arsenicales/farmacología , Células Sanguíneas/citología , Células Sanguíneas/metabolismo , Recolección de Muestras de Sangre , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Ratones , Óxidos/farmacología
10.
J Stem Cells ; 5(2): 49-64, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-22049615

RESUMEN

Myelodysplastic syndromes (MDSs) represent a spectrum of disorders that are generally thought to arise from a defective hematopoietic stem cell leading to clonal, dysregulated hematopoiesis. Although it is generally agreed that the marrow microenvironment plays a role in the biology of MDS, it is unclear whether this represents an intrinsically abnormal stromal compartment derived from the MDS clone. Hematopoiesis requires cooperation between progenitors and a variety of functionally and phenotypically different cell types that form the bone marrow stroma. Stromal abnormalities suspected to contribute to the pathology of bone marrow disorder with impaired hematopoiesis. Several studies on human MDS bone marrow microenvironment revealed functional alteration and increased cellular apoptosis thus contribute to the pathology of the disease progression. In this present study, we have investigated alterations in the hematopoietic microenvironment and underlying mechanisms involved in the disease progression of MDS animal model. We presented the results of bone marrow single cell culture study, Long-term bone marrow adherent culture study (LTBMC) and their functional efficacy, flowcytometric characterization of stem (Scal+c-kit+) and stromal (Scal+CD44+) progenitor cell population and expression level of extracellular apoptosis marker (Annexin v) in the bone marrow cells of MDS animal model. Bone marrow single cell culture study of MDS animal showed impairment in the normal cellular generation, proliferation and presence of apoptic cells. Long-term liquid Bone marrow stromal cell colony formation assay from MDS bone marrow cells showed significant difference in the colony formation and their maintenance than the control groups of animals. Immune functional capacity of the bone marrow stromal cells through cell mediated immune (CMI) parameter study denoted defects in the stromal microenvironment. Decreased expression of bone marrow long-term primitive hematopoietic population and stromal progenitor population depicted bone marrow abnormality in case of MDS animal model, which bears significant correlation with high expression level of apoptosis marker in the bone marrow cells. From the above experimental study we tried to highlight the abnormal bone marrow microenvironment and alteration in the bone marrow cell surface marker expression, which could be the probable mechanism of evolution and disease progression in case of MDS animal model.


Asunto(s)
Médula Ósea/patología , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/patología , Células Madre Mesenquimatosas/patología , Síndromes Mielodisplásicos/patología , Células del Estroma/patología , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Humanos , Masculino , Ratones
11.
Curr Stem Cell Res Ther ; 5(4): 379-86, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20528754

RESUMEN

Leukemia is a heterogeneous disorder of bone marrow (BM) failure syndrome where normal hematopoiesis gets altered due to transformation of either the normal hematopoietic cell or the hematopoietic microenvironment or both. Scientists have tried for decades to understand leukemia development in the context of therapeutic strategies. The existence of "leukemic stem cells" and their possible role in leukemogenesis have only recently been identified and it has changed the perspective with regard to new approaches for treating the disease. However the relationship between leukemic stem cells (LSCs) and leukemogenesis requires further investigation. In this present study, we have experimentally induced leukemia in mice by means of N-N' Ethylnitrosourea (ENU) to investigate the alterations in normal bone marrow cellular phenotype and associated changes in the stromal hematopoietic microenvironment under the event of leukemic disease progression. We have identified a significant decrease in the normal HSC phenotype in terms of Sca1 and c-kit receptor expression and subsequent sharp increase in certain leukemic cell specific receptor expression like CD123, CXCR4 and CD44 in the leukemic bone marrow. The decreased HSC receptor (Sca1 and c-kit) expression profile with concurrent increase in the expression of leukemic cell specific receptors (CD123, CXCR4, CD44) by the bone marrow cells of leukemic mice may account for the possible transformation of the normal hematopoietic cells that is necessary for the disease initiation and progression. Some of these receptors like CXCR4 and CD44 are also known to play an important role in maintaining leukemic cells and their complex crosstalk with the surrounding stromal microenvironment. Thus up-regulation in CXCR4 and CD44 receptor expression essentially pointed towards the stroma dependent surveillance of the leukemic bone marrow cells in leukemia. Leukemic bone marrow cells documented a rapid generation of stromal feeder layer in culture. The rapid stroma generation further supported the fact that leukemic stromal microenvironment gets altered in possible ways to support leukemic cell generation and fueling leukemogenesis. The study presented here, has tried to hint at exploring new therapeutic strategies by not only identifying the expression profile of cell surface receptors unique to cells involved in leukemic progression but also targeting the specific components of the stromal microenvironment that would facilitate therapeutic management of the disease.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Leucemia/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Médula Ósea/patología , Transformación Celular Neoplásica/inducido químicamente , Células Cultivadas , Progresión de la Enfermedad , Etilnitrosourea/administración & dosificación , Células Madre Hematopoyéticas/patología , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Inmunofenotipificación , Leucemia/inducido químicamente , Leucemia/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Microambiente Tumoral/fisiología
12.
Stem Cells Int ; 2010: 614395, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21048851

RESUMEN

Self-renewing Hematopoietic Stem Cells (HSCs) are responsible for reconstitution of all blood cell lineages. Sca-1 is the "stem cell antigen" marker used to identify the primitive murine HSC population, the expression of which decreases upon differentiation to other mature cell types. Sca-1(+) HSCs maintain the bone marrow stem cell pool throughout the life. Aplastic anemia is a disease considered to involve primary stem cell deficiency and is characterized by severe pancytopenia and a decline in healthy blood cell generation system. Studies conducted in our laboratory revealed that the primitive Sca-1(+) BM-HSCs (bone marrow hematopoietic stem cell) are significantly affected in experimental Aplastic animals pretreated with chemotherapeutic drugs (Busulfan and Cyclophosphamide) and there is increased Caspase-3 activity with consecutive high Annexin-V positivity leading to premature apoptosis in the bone marrow hematopoietic stem cell population in Aplastic condition. The Sca-1(bright), that is, "more primitive" BM-HSC population was more affected than the "less primitive" BM-HSC Sca-1(dim ) population. The decreased cell population and the receptor expression were directly associated with an empty and deranged marrow microenvironment, which is evident from scanning electron microscopy (SEM). The above experimental evidences hint toward the manipulation of receptor expression for the benefit of cytotherapy by primitive stem cell population in Aplastic anemia cases.

13.
Stem Cells Int ; 2010: 932354, 2010 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21048856

RESUMEN

Aplastic anemia (AA) is a heterogeneous disorder of bone marrow failure syndrome. Suggested mechanisms include a primary stem cell deficiency or defect, a secondary stem cell defect due to abnormal regulation between cell death and differentiation, or a deficient microenvironment. In this study, we have tried to investigate the alterations in hematopoietic microenvironment and underlying mechanisms involved in such alterations in an animal model of drug induced AA. We presented the results of studying long term marrow culture, marrow ultra-structure, marrow adherent and hematopoietic progenitor cell colony formation, flowcytometric analysis of marrow stem and stromal progenitor populations and apoptosis mechanism involved in aplastic anemia. The AA marrow showed impairment in cellular proliferation and maturation and failed to generate a functional stromal microenvironment even after 19 days of culture. Ultra-structural analysis showed a degenerated and deformed marrow cellular association in AA. Colony forming units (CFUs) were also severely reduced in AA. Significantly decreased marrow stem and stromal progenitor population with subsequently increased expression levels of both the extracellular and intracellular apoptosis inducer markers in the AA marrow cells essentially pointed towards the defective hematopoiesis; moreover, a deficient and apoptotic microenvironment and the microenvironmental components might have played the important role in the possible pathogenesis of AA.

14.
Int J Stem Cells ; 3(1): 54-62, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-24855541

RESUMEN

The wide use of pesticides for agriculture, domestic and industrial purposes and evaluation of their subsequent effect is of major concern for public health. Human exposure to these contaminants especially bone marrow with its rapidly renewing cell population is one of the most sensitive tissues to these toxic agents represents a risk for the immune system leading to the onset of different pathologies. In this experimental protocol we have developed a mouse model of pesticide(s) induced hypoplastic/aplastic marrow failure to study quantitative changes in the bone marrow hematopoietic stem cell (BMHSC) population through flowcytometric analysis, defects in the stromal microenvironment through short term adherent cell colony (STACC) forming assay and immune functional capacity of the bone marrow derived cells through cell mediated immune (CMI) parameter study. A time course dependent analysis for consecutive 90 days were performed to monitor the associated changes in the marrow's physiology after 30(th), 60(th) and 90(th) days of chronic pesticide exposure. The peripheral blood showed maximum lowering of the blood cell count after 90 days which actually reflected the bone marrow scenario. Severe depression of BMHSC population, immune profile of the bone marrow derived cells and reduction of adherent cell colonies pointed towards an essentially empty and hypoplastic marrow condition that resembled the disease aplastic anemia. The changes were accompanied by splenomegaly and splenic erythroid hyperplasia. In conclusion, this animal model allowed us a better understanding of clinico-biological findings of the disease aplastic anemia following toxic exposure to the pesticide(s) used for agricultural and industrial purposes.

15.
J Stem Cells ; 5(2): 65-74, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-22049616

RESUMEN

Stem Cell Antigen-1 or Sca-1 is a cell surface receptor protein commonly used to detect adult murine haematopoietic stem cell population. Outside the haematopoietic system Sca-1 is similarly expressed in stem and progenitor cells in a wide variety of tissues and organs such as skeletal muscle, mammary gland, prostate, heart, liver and dermis. Thus Sca-1 has become a candidate marker in the search of tissue specific stem cells. The maintenance of a healthy corneal epithelium is achieved by a unique population of stem cell located in the limbal epithelial region. This limbal epithelium mainly contains limbal epithelial stem cells and its immediate progenitor early transient amplifying cells (e-TAC) which have self renewal capacity. As stem cells in other organs have been identified by their expression of Sca-1, in our study we wanted to determine whether this antigen could be present in the limbal epithelial region which contains stem cell population by using immunofluorescence through flow cytometric analysis of Sca-1 and its association with the cell cycle.


Asunto(s)
Antígenos Ly/metabolismo , Ciclo Celular/fisiología , Epitelio Corneal/metabolismo , Limbo de la Córnea/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Dactinomicina/análogos & derivados , Epitelio Corneal/citología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes , Humanos , Limbo de la Córnea/citología , Masculino , Ratones , Células Madre/citología
16.
J Stem Cells ; 4(3): 179-89, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20232602

RESUMEN

The production of blood cells from bone marrow (BM) hematopoietic stem cells (HSC) is regulated by a number of cytokines and growth factors that influence cell survival; differentiation, proliferation and apoptosis in health and supposedly, such mechanisms are deregulated in diseased conditions. As far as cellular kinetics is concerned HSCs are relatively quiescent in adults, have the ability to replicate symmetrically and asymmetrically and predictably exhibit multi-lineage hematopoietic reconstitution potential. HSC drive hematopoiesis and homeostasis by contracting and expanding the pool of hematopoietic cells in the bone marrow. In mouse they can be identified immunophenotypically as Sca1+ c-kit cells. In aplastic anemia a drastic decline in the marrow efficacy to produce mature blood cells leads to bone marrow failure. In contrast, in leukemia hyper stimulated marrow leads to deregulated differentiation of immature hematopoietic stem cells with increased self-proliferation potential. In our experimental set up, we induced aplastic anemia by injecting busulfan and cyclophosphamide and leukemia by N-N' ethylnitrosourea intraperitoneally in inbred swiss albino mice. Indeed, HSCs and haematopoietic progenitor cells (HPCs) are vulnerable target for such disease oriented dysregulation which bears close correlation with the bone marrow microenvironmental damage. The present study aims at evaluating the possible mechanism(s) of deregulation in the bone marrow physiology with special reference to HSC surface receptor expression, cellular granularity, cell cycle status and overall marrow architecture. The investigations made so far revealed an interesting correlation between disease initiation and specific cytokinetic involvement of HSC in the BM microenvironment with particular reference to leukemia and aplastic anemia.


Asunto(s)
Anemia Aplásica/patología , Células de la Médula Ósea/patología , Proliferación Celular , Células Madre Hematopoyéticas/patología , Leucemia Experimental/patología , Anemia Aplásica/inducido químicamente , Anemia Aplásica/metabolismo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/metabolismo , Busulfano , Ciclo Celular , Ciclofosfamida , Etilnitrosourea , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Cinética , Leucemia Experimental/inducido químicamente , Leucemia Experimental/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo
17.
J Stem Cells ; 4(4): 229-41, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20720596

RESUMEN

The bone marrow is the major site of haemopoiesis in adult human. It contains cells that represent the stages in the development of different types of blood cells e.g. myelocytes, metamyelocytes, erythroblasts, reticulocytes, and other lymphoid progenies etc. Bone marrow failure is primarily the result of a specific failure of bone marrow precursor cells to produce mature cells. N-ethyl N-nitroso urea (ENU) is one of the most potent mutagens that can create an abnormal bone marrow microenvironment by causing defect in haematopoietic stem cell maturation cascade. ENU is easy to administer in mouse, and some probable mutations can be helpful to create models of human diseased conditions like Myelodysplastic syndrome (MDS). MDS is considered as an intravascular bone marrow disorder, a combined structural-functional abnormality wherein the differentiation procedure of the bone marrow stem cell is either incomplete or defective. We assumed that Myelodysplastic syndrome stands in between an inhibitory cellular pattern and a positive overshoot of abnormal differentiations representing an unknown juncture where the mystery of aplasia and leukemia hide back. Instead of using a transgenic mouse model, we attempted to develop an experimentally induced murine model of preleukemia or human MDS like disease model. In doing so ENU has been administered i.p and the animals were examined on thirtieth day and peripheral blood haemogram was documented. Upon registering the appearance of abnormal peripheral blood scenario, the changes in the intravascular bone marrow (BM) architecture, cell surface receptor expression, e.g. Sca-1, c-Kit and the early and late phase apoptic patterns were noted. The results represented an interesting correlation in between bone marrow architecture, early stem cell receptor and apoptic marker expression resembling human MDS.


Asunto(s)
Antígenos Ly/metabolismo , Apoptosis/fisiología , Proteínas de la Membrana/metabolismo , Síndromes Mielodisplásicos/inducido químicamente , Síndromes Mielodisplásicos/metabolismo , Compuestos de Nitrosourea/toxicidad , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Antígenos Ly/genética , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Citometría de Flujo , Humanos , Proteínas de la Membrana/genética , Ratones , Síndromes Mielodisplásicos/fisiopatología , Proteínas Proto-Oncogénicas c-kit/genética , Células Madre/citología , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA