Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
World J Stem Cells ; 12(10): 1050-1066, 2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33178391

ABSTRACT

Inflammatory bowel disease (IBD), consisting primarily of ulcerative colitis and Crohn's disease, is a group of debilitating auto-immune disorders, which also increases the risk of colitis-associated cancer. However, due to the chronic nature of the disease and inconsistent treatment outcomes of current anti-IBD drugs (e.g., approximately 30% non-responders to anti-TNFα agents), and related serious side effects, about half of all IBD patients (in millions) turn to alternative treatment options. In this regard, mucosal healing is gaining acceptance as a measure of disease activity in IBD patients as recent studies have correlated the success of mucosal healing with improved prognosis. However, despite the increasing clinical realization of the significance of the concept of mucosal healing, its regulation and means of therapeutic targeting remain largely unclear. Here, stem-cell therapy, which uses hematopoietic stem cells or mesenchymal stem cells, remains a promising option. Stem cells are the pluripotent cells with ability to differentiate into the epithelial and/or immune-modulatory cells. The over-reaching concept is that the stem cells can migrate to the damaged areas of the intestine to provide curative help in the mucosal healing process. Moreover, by differentiating into the mature intestinal epithelial cells, the stem cells also help in restoring the barrier integrity of the intestinal lining and hence prevent the immunomodulatory induction, the root cause of the IBD. In this article, we elaborate upon the current status of the clinical management of IBD and potential role of the stem cell therapy in improving IBD therapy and patient's quality of life.

2.
Curr Med Chem ; 27(37): 6276-6293, 2020.
Article in English | MEDLINE | ID: mdl-31584360

ABSTRACT

BACKGROUND: Chronic Liver Disorders (CLD), caused by the lifestyle patterns like alcoholism or by non-alcoholic fatty liver disease or because of virus-mediated hepatitis, affect a large population fraction across the world. CLD progresses into end-stage diseases with a high mortality rate. Liver transplant is the only approved treatment available for such end-stage disease patients. However, the number of liver transplants is limited due to the limited availability of suitable donors and the extremely high cost of performing the procedure. Under such circumstances, Stem Cell (SC) mediated liver regeneration has emerged as a potential therapeutic alternative approach. OBJECTIVE: This review aims to critically analyze the current status and future prospects of stem cellbased interventions for end-stage liver diseases. The clinical studies undertaken, the mechanism underlying therapeutic effects and future directions have been examined. METHOD: The clinical trial databases were searched at https://clinicaltrials.gov.in and http://www.isrctn.com to identify randomized, non-randomized and controlled studies undertaken with keywords such as "liver disorder and Mesenchymal Stem Cells (MSCs)", "liver cirrhosis and MSCs" and "liver disorder and SCs". Furthermore, https://www.ncbi.nlm.nih.gov/pubmed/ database was also explored with similar keywords for finding the available reports and their critical analyses. RESULTS: The search results yielded a significant number of studies that used bone marrow-derived stem cells, MSCs and hepatocytes. The studies clearly indicated that SCs play a key role in the hepatoprotection process by some mechanisms involving anti-inflammation, auto-immune-suppression, angiogenesis and anti-apoptosis. Further, studies indicated that SCs derived paracrine factors promote angiogenesis, reduce inflammation and inhibit hepatocyte apoptosis. CONCLUSION: The SC-based interventions provide a significant improvement in patients with CLD; however, there is a need for randomized, controlled studies with the analysis of a long-term follow-up.


Subject(s)
Liver , Mesenchymal Stem Cells , Hepatocytes , Humans , Liver Diseases/therapy , Liver Regeneration , Mesenchymal Stem Cell Transplantation
3.
Stem Cells Int ; 2013: 496218, 2013.
Article in English | MEDLINE | ID: mdl-23577036

ABSTRACT

Mesenchymal stem cells (MSCs) are stromal cells that have the ability to self-renew and also exhibit multilineage differentiation into both mesenchymal and nonmesenchymal lineages. The intrinsic properties of these cells make them an attractive candidate for clinical applications. MSCs are of keen interest because they can be isolated from a small aspirate of bone marrow or adipose tissues and can be easily expanded in vitro. Moreover, their ability to modulate immune responses makes them an even more attractive candidate for regenerative medicine as allogeneic transplant of these cells is feasible without a substantial risk of immune rejection. MSCs secrete various immunomodulatory molecules which provide a regenerative microenvironment for a variety of injured tissues or organ to limit the damage and to increase self-regulated tissue regeneration. Autologous/allogeneic MSCs delivered via the bloodstream augment the titers of MSCs that are drawn to sites of tissue injury and can accelerate the tissue repair process. MSCs are currently being tested for their potential use in cell and gene therapy for a number of human debilitating diseases and genetic disorders. This paper summarizes the current clinical and nonclinical data for the use of MSCs in tissue repair and potential therapeutic role in various diseases.

4.
Stem Cells Dev ; 22(12): 1802-17, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23343006

ABSTRACT

Pluripotency and stemness is believed to be associated with high Oct-3/4, Nanog, and Sox-2 (ONS) expression. Similar to embryonic stem cells (ESCs), high ONS expression eventually became the measure of pluripotency in any cell. The threshold expression of ONS genes that underscores pluripotency, stemness, and differentiation potential is still unclear. Therefore, we raised a question as to whether pluripotency and stemness is a function of basal ONS gene expression. To prove this, we carried out a comparative study between basal ONS expressing NIH3T3 cells with pluripotent mouse bone marrow mesenchymal stem cells (mBMSC) and mouse ESC. Our studies on cellular, molecular, and immunological biomarkers between NIH3T3 and mBMSC demonstrated stemness property of undifferentiated NIH3T3 cells that was similar to mBMSC and somewhat close to ESC as well. In vivo teratoma formation with all three germ layer derivatives strengthen the fact that these cells in spite of basal ONS gene expression can differentiate into cells of multiple lineages without any genetic modification. Conclusively, our novel findings suggested that the phenomenon of pluripotency which imparts ability for multilineage cell differentiation is not necessarily a function of high ONS gene expression.


Subject(s)
Bone Marrow Cells/metabolism , Embryonic Stem Cells/metabolism , Gene Expression , Germ Layers/metabolism , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Survival , Embryonic Stem Cells/cytology , Germ Layers/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Primary Cell Culture , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Teratoma/metabolism , Teratoma/pathology
5.
Curr Top Med Chem ; 11(13): 1592-8, 2011.
Article in English | MEDLINE | ID: mdl-21446911

ABSTRACT

Normal tissue homeostasis involves a careful balance between the normal cell loss and renewal. Stem and progenitor cells help maintain this precise and fine balance through their ability of self-renewal in a tightly regulated manner. In this regard, the gastrointestinal epithelium is unique in that cell proliferation, differentiation, and apoptosis occur in an orderly fashion along the crypt-villus axis. The colonic crypt is primarily a proliferative compartment, is monoclonal and is maintained by stem cells. The concept of tissue stem cells capable of giving rise to all differentiated cells within a given tissue has led to the concept of a cellular hierarchy in tissues and in tumors including colorectal cancer (CRC). Thus, only a few cells may be necessary and sufficient for tissue repair or tumor regeneration. However, such a proposition also raises questions regarding the precise methods and markers to identify such population and to define the circumstantial evidences and place for the origin and establishment of the early mutant stem-cell population. Thus, it is imperative that we understand what cancer stem cells (CSC) are and their potential association with cancer in a tissue specific manner. In this review, we have summarized the current knowledge of stem cell organization and CSC within the colonic epithelium and discussed the potential role of CSC in the development and/or progression of CRC and as targets for therapeutic interventions.


Subject(s)
Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Cell Transformation, Neoplastic/pathology , Humans , Intestinal Mucosa/pathology
7.
Stem Cells ; 29(3): 486-95, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21425411

ABSTRACT

Pluripotent stem cells hold significant promise in regenerative medicine due to their unlimited capacity for self-renewal and potential to differentiate into any cell type of the body. In this study, we demonstrate that proper mitochondrial function is essential for proliferation of undifferentiated ESCs. Attenuating mitochondrial function under self-renewing conditions makes these cells more glycolytic-dependent, and it is associated with an increase in the mRNA reserves of Nanog, Oct4, and Sox2. In contrast, attenuating mitochondrial function during the first 7 days of differentiation results in normal repression of Oct4, Nanog, and Sox2. However, differentiation potential is compromised as revealed by abnormal transcription of multiple Hox genes. Furthermore, under differentiating conditions in which mitochondrial function is attenuated, tumorigenic cells continue to persist. Our results, therefore establish the importance of normal mitochondrial function in ESC proliferation, regulating differentiation, and preventing the emergence of tumorigenic cells during the process of differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/physiology , Mitochondria/physiology , Animals , Cell Culture Techniques , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, SCID , Pluripotent Stem Cells/physiology , Time Factors
8.
J Transl Med ; 7: 27, 2009 Apr 22.
Article in English | MEDLINE | ID: mdl-19386101

ABSTRACT

BACKGROUND: It has been suggested that the initial differentiation of endothelial and hematopoietic cells during embryogenesis occurs from a common progenitor, called hemangioblast (hB). We hypothesized that these cells with dual hematopoietic/endothelial potential could be used in future regenerative medicine. METHODS: We used the two-step differentiation technology to generate bipotential blast cells from human embryonic stem cells (hES). This involved short differentiation in our in vitro EB system followed by differentiation in semisolid culture medium supplemented with mixture of cytokines. RESULTS: The occurrence of blast-colony-forming cells (BL-CFC) during EB differentiation (day 0-6) was transient and peaked on day 3. The emergence of this event was associated with expression of mesoderm gene T, and inversely correlated with expression of endoderm gene FoxA2. Similarly, the highest BL-CFC number was associated with increase in expression of early hematopoietic/endothelial genes: CD34, CD31 and KDR. The derived colonies were composed of 30-50 blast cells on day 6 in culture. These cells had homogenous appearance in Wright-Giemsa stain, but to a different extent expressed markers of immature hematopoietic and endothelial cells (CD31, CD34, VE-cadherin, Flt-1) and mature differentiated cells (CD45, CD33, CD146). We found that some of them expressed fetal and embryonic globin genes. Interestingly, these cells expressed also HLA class I molecules, however at very low levels compared to endothelial and hematopoietic cells. The blast cells could be successfully differentiated to hematopoietic cells in a CFU assay. In these conditions, blast cells formed CFU-M colonies (63.4 +/- 0.8%) containing macrophages, BFU-E colonies (19.5 +/- 3.5%) containing nucleated red blood cells, and CFU-EM colonies (17.1 +/- 2.7%) composed of macrophages and nucleated erythrocytes. Cells of CFU-EM and BFU-E colonies expressed both epsilon - and gamma- globin genes, but not adult-type gamma-globin. When in endothelial cell culture conditions, blast cells differentiated to endothelial cells which had the ability to take up Dil-Ac-LDL and to form complex vascular networks in Matrigel. CONCLUSION: 1) Hematoendothelial precursors exist transiently in early embryonic development and form single cell-derived colonies; 2) their differentiation can be tracked by the use of chosen molecular markers; 3) blast colonies consist of cells having properties of endothelial and hematopoietic precursors, however the issue of their ability to maintain dual properties over time needs to be further explored; 4) blast cells can potentially be used in regenerative medicine due to their low expression of HLA molecules.


Subject(s)
Embryonic Stem Cells/immunology , HLA-A Antigens/genetics , Hemangioblasts/immunology , Blood Vessels/embryology , Blood Vessels/growth & development , Cell Differentiation , Cell Division , Collagen , Drug Combinations , Embryonic Development , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Endothelial Cells/physiology , Fluorescent Antibody Technique , Hemangioblasts/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Laminin , Proteoglycans
9.
Curr Pharm Biotechnol ; 10(3): 345-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19355944

ABSTRACT

"Niche" is defined as a specialized regulatory microenvironment, consisting of components which control the fate specification of stem and progenitor cells, as well as maintaining their development by supplying the requisite factors. Bone marrow (BM) niche has a well-organized architecture and is composed of osteoblasts, osteoclasts, bone marrow endothelial cells, stromal cells, adipocytes and extracellular matrix proteins (ECM). These elements play an essential role in the survival, growth and differentiation of diverse lineages of blood cells, but also provide optimal growth environment for multiple hematological malignancies including multiple myeloma (MM). MM is a neoplastic plasma cell disorder which not only resides in BM but also converts it into specialized neoplastic niche. This niche aids the growth and spreading of tumor cells by a complex interplay of cytokines, chemokines, proteolytic enzymes and adhesion molecules. Moreover, the MM BM microenvironment was shown to confer survival and chemoresistance of MM cells to current therapies. However, our knowledge in this field is still in infancy and many details are unknown. Therefore, there is a strong need to further dissect the MM BM niche and understand the process of how the complex interactions with BM milieu influence MM growth, survival and development of resistance to chemotherapy. A better and more detailed understanding of neoplastic MM niche will provide a guiding model for identifying and validating novel targeted therapies directed against MM. Therefore, in the present review, we have focused principally on the basic features, physical structures, and functions of the BM niche and have highlighted its interaction with MM cells.


Subject(s)
Bone Marrow/pathology , Multiple Myeloma/pathology , Adipocytes/physiology , Animals , Dendritic Cells/physiology , Endothelial Cells/physiology , Extracellular Matrix/pathology , Humans , Osteoblasts/physiology , Osteoclasts/physiology , Stromal Cells/metabolism , Stromal Cells/pathology
10.
Curr Pharm Des ; 14(36): 3873-9, 2008.
Article in English | MEDLINE | ID: mdl-19128238

ABSTRACT

Neurodegenerative diseases comprise a heterogeneous spectrum of neural disorders that cause severe and progressive cognitive and motor deficits. A histological hallmark of these disorders is the occurrence of disease-specific cell death in specific regional subpopulations of neurons, such as the loss of dopaminergic neurons in the substantia nigra in Parkinson's disease. Neurodegenerative disease can also possibly occur from the loss or dysfunction of selected glial cell subsets, such as the dysfunction of supportive glial cells around somatic motor neurons in amyotrophic lateral sclerosis. The central nervous system (CNS), unlike many other tissues, has a very limited capacity for self-repair. Mature nerve cells lack the ability to regenerate, although endogenous neural stem cells exist in the adult brain that do have very limited ability to generate new functional neurons in response to injury. Rapid advances in stem cell biology have opened an alternative, fascinating perspective of neurogenesis by activation of endogenous neural stem cells and/or transplantation of in vitro-expanded stem cells and/or their neuronal- or glial-differentiated progeny. Embryonic stem (ES) cells, because of their ability to provide seemingly unlimited supply of specific cell types, their amenability to genetic engineering manipulations, and their broad developmental potential, are expected to become a cell source and biological delivery system for use in a variety of neurodegenerative diseases, and are likely to play a role in the development of novel cell-based therapies for these indications. However, before the full potential of ES cells can be realized for regenerative medicine, we need to understand mechanisms regulating their proliferation, differentiation into therapeutically relevant cells, and most importantly in the case of neuronal and glial lineages, to characterize their functional properties. In the present review we will be focusing on the factors and methodologies responsible for differentiation of ES cell into different neural precursors and neural cell lineages with particular emphasis on the potential research and clinical applications of ES cells in the field of neurodegenerative disease.


Subject(s)
Embryonic Stem Cells , Neurodegenerative Diseases/therapy , Animals , Cell Transplantation , Humans , Mice , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/therapeutic use , Neurodegenerative Diseases/drug therapy
11.
Curr Pharm Biotechnol ; 8(5): 305-17, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17979728

ABSTRACT

Cellular therapies derived from embryonic stem (ES) cells have gained a renewed interest with the experimental demonstration that an embryonic stem cell lines can be established from human blastocyst-stage embryos and prompted to differentiate into almost all types of cells present in the body including hematopoietic cells. Hematopoiesis is a series of cellular processes whereby short-lived mature blood cells are continuously replenished from a pool of rare pluripotential hematopoietic stem cells, in a highly orchestrated process. Aberrances in this intricate process may lead to a malignancy of essential blood-forming organs, causing diseases such as leukemia, aplastic anemia, lymphoma, myelodysplasia and myeloproliferative disorders. Embryonic stem cells show great potential and it may be technologically feasible to transplant differentiated ES cells and to cure various kinds of blood disorders. Understanding the biology of ES cell derived hematopoiesis may lead to the development of co-transplantation protocols that will result in a decreased morbidity and mortality by providing safer and simpler transplantation procedures for patients with malignant and non-malignant conditions. The potential utility of ES cells for gene therapy, tissue engineering and the treatment of a wide variety of currently untreatable diseases is simply too essential to ignore, however, our knowledge and ability to deliver these forms of therapy in a safe and efficient manner requires additional advances in the understanding of the basic biology of ES cells. In this article, we will discuss the factors and methodologies responsible for the differentiation of ES cells into hematopoietic progenitors and their potential to treat different blood related diseases.


Subject(s)
Embryonic Stem Cells/transplantation , Hematologic Diseases/surgery , Hematopoietic Stem Cell Transplantation/trends , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Hematologic Diseases/pathology , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans
12.
Biochem Biophys Res Commun ; 361(4): 953-9, 2007 Oct 05.
Article in English | MEDLINE | ID: mdl-17692287

ABSTRACT

The primary objective of this work is to determine the repairing potential of murine embryonic stem cells (ES) in murine model of Crohn's disease (CD). Colitis, induced in IL10-/- KO mice using piroxicam, was associated with the increased levels of IL-12. Enhanced yellow fluorescent protein (EYFP) marked murine ES cells (R1/129) and control non-fluorescent ES cells were subjected to in vitro differentiation into intestinal epithelial cells. IL 10-/- KO mice were injected with pre-differentiated ES-YFP cells and sacrificed after 2 and 3 months. Histopathological analysis of intestines demonstrated a progressive improvement in colitis (from grade-4 to grade-1 and -0) and decreased levels of IL-12 cytokine following transplantation. Fluorescent and confocal microscopy demonstrated presence of ES-EYFP cells in the colon, small intestine, liver, and thymus tissues but none in the spleen and bone marrow. The EYFP signal was not detected in sham (non-transplanted mice with induced colitis) and control IL10-/- KO mice. Engraftment, detected at 3 months post-transplant, correlated with markedly improved grading in colon histology (grade-1 or -0) and weight gain, as well as with decreased rectal prolapses. In vitro pre-differentiated ES cells migrated and homed exclusively into the colon, small intestine, and the liver, engrafted for long term, reduced inflammation and tissue damage, and restored immune balance. These findings suggest that pre-differentiated ES cells may become alternative source of stem cell therapy for CD with dual functions i.e. regenerating damaged epithelium and restoring immune imbalance occurring in this disease.


Subject(s)
Crohn Disease/therapy , Embryonic Stem Cells/transplantation , Interleukin-10/genetics , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/therapy , Colon/pathology , Crohn Disease/chemically induced , Crohn Disease/immunology , Interleukin-10/metabolism , Mice , Mice, Knockout , Piroxicam
13.
Stem Cells ; 25(6): 1456-61, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17379761

ABSTRACT

The role of thrombopoietin (TPO) in adult hematopoiesis is well-established. A recent report suggests that TPO and vascular endothelial growth factor (VEGF) play a role in promoting formation of early erythropoietic progenitors in a nonhuman primate embryonic stem cell (ES) model. No such report exists for human ES cells as yet. Because TPO may become an important factor promoting human ES cell-derived hematopoiesis, we sought to investigate whether TPO in combination with VEGF can enhance human ES-derived hematopoiesis in an EB-derived culture system. The emphasis of this work was to demonstrate the molecular mechanisms involved in this process, specifically the role of c-mpl and its ligand TPO. Human ES cells were cultured to the EB state, and EB-derived secondary cultures supporting hematopoietic differentiation were established: condition 1, control (stem cell factor [SCF] and Flt3 ligand [Flt3L]); condition 2, SCF, Flt3L, and TPO; and condition 3, SCF, Flt3L, TPO, and VEGF. Cells were harvested daily, starting at day 2 and continuing until day 8, for reverse transcription-polymerase chain reaction and Western blot. There was no evidence of expression of c-mpl and VEGF receptor on the gene or protein level until day 8, when the formation of well-established hematopoietic colonies began. This correlated with the formation of CD34+/CD31- negative progenitors, mostly found in blast-forming units-erythroid-like colonies. We concluded that TPO and VEGF play an important synergistic role in the formation of early ES-derived hematopoietic progenitors that occurs through the c-mpl and VEGF receptors. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Thrombopoietin/pharmacology , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/cytology , Humans , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Thrombopoietin/physiology
14.
Biochem Biophys Res Commun ; 346(2): 508-16, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16764825

ABSTRACT

Differentiating embryonic stem (ES) cells are increasingly emerging as an important source of hematopoietic progenitors with a potential to be useful for both basic and clinical research applications. It has been suggested that dexamethasone facilitates differentiation of ES cells towards erythrocytes but the mechanism responsible for sequential expression of genes regulating this process are not well-understood. Therefore, we in vitro induced differentiation of murine ES cells towards erythropoiesis and studied the sequential expression of a set of genes during the process. We hypothesized that dexamethasone-activates its cognate nuclear receptors inducing up-regulation of erythropoietic genes such as GATA-1, Flk-1, Epo-R, and direct ES cells towards erythropoietic differentiation. ES cells were cultured in primary hematopoietic differentiation media containing methyl-cellulose, IMDM, IL-3, IL-6, and SCF to promote embryoid body (EB) formation. Total RNA of day 3, 5, and 9-old EBs was isolated for gene expression studies using RT-PCR. Cells from day 9 EBs were subjected to secondary differentiation using three different cytokines and growth factors combinations: (1) SCF, EPO, dexamethasone, and IGF; (2) SCF, IL-3, IL-6, and TPO; and, (3) SCF IL-3, IL-6, TPO, and EPO. Total RNA from day 12 of secondary differentiated ES cells was isolated to study the gene expression pattern during this process. Our results demonstrate an up-regulation of GATA-1, Flk-1, HoxB-4, Epo-R, and globin genes (alpha-globin, betaH-1 globin, beta-major globin, epsilon -globin, and zeta-globin) in the 9-day-old EBs, whereas, RNA from 5-day-old EBs showed expression of HoxB-4, epsilon-globin, gamma-globin, betaH1-globin, and Flk-1. Three-day-old EBs showed only HoxB-4 and Flk-1 gene expression and lacked expression of all globin genes. These findings indicate that erythropoiesis-specific genes are activated later in the course of differentiation. Gene expression studies on the ES cells of secondary EB origin cultured in media containing dexamethasone showed a down-regulation of GATA-3 and an up-regulation of GATA-1, Flk-1, and Epo-R in comparison to the two other cytokines and growth factor combinations containing media. The secondary differentiation also showed an enhanced production of erythrocytic precursors in dexamethasone containing media in comparison to that in the control media. Our results indicate that dexamethasone can prove to be an effective agent which can be employed to enhance differentiation towards erythrocytic progenitors from ES cells.


Subject(s)
Cell Differentiation , Dexamethasone/pharmacology , Erythropoiesis/drug effects , Glucocorticoids/pharmacology , Pluripotent Stem Cells/drug effects , Animals , Cells, Cultured , Culture Media , Embryo, Mammalian/cytology , Erythropoiesis/genetics , Erythropoietin/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Stem Cell Factor/pharmacology
15.
Stem Cells ; 24(7): 1689-94, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574752

ABSTRACT

An understanding of feasibility of implanting embryonic stem cells (ESCs), their behavior of migration in response to lesions induced in brain tissues, and the mechanism of their in vivo differentiation into neighboring neural cells is essential for developing and refining ESC transplantation strategies for repairing damages in the nervous system, as well as for understanding the molecular mechanism underlying neurogenesis. We hypothesized that damaged neural tissues offer a niche to which injected ESCs can migrate and differentiate into the neural cells. We inflicted damage in the murine (C57BL/6) brain by injecting phosphate-buffered saline into the left frontal and right caudal regions and confirmed neural damage by histochemistry. Enhanced yellow fluorescent protein-expressing ESCs were injected into the nondamaged left caudal portion of the brain. Using immunohistochemistry and fluorescent microscopy, we observed migration of ESCs from the injection site (left caudal) to the damaged site (right caudal and left frontal). Survival of the injected ESCs was confirmed by the real-time polymerase chain reaction analysis of stemness genes such as Oct4, Sox2, and FGF4. The portions of the damaged neural tissues containing ESCs demonstrated a fourfold increase in expression of these genes after 1 week of injection in comparison with the noninjected ESC murine brain, suggesting proliferation. An increased level of platelet-derived growth factor receptor demonstrated that ESCs responded to damaged neural tissues, migrated to the damaged site of the brain, and proliferated. These results demonstrate that undifferentiated ESCs migrate to the damaged regions of brain tissue, engraft, and proliferate. Thus, damaged brain tissue provides a niche that attracts ESCs to migrate and proliferate.


Subject(s)
Brain Damage, Chronic/therapy , Cell Movement , Cell Proliferation , Cell Survival , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Buffers , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 4/metabolism , Gene Expression , Head Injuries, Penetrating/chemically induced , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors , Trans-Activators/metabolism
16.
Article in English | MEDLINE | ID: mdl-20483231

ABSTRACT

Alpha 1 chain (Colalpha1(I)) and alpha 2 chain (Colalpha2(I)) are universal components of type I collagen in tetrapods, but rainbow trout (Oncorhynchus mykiss) and zebrafish (Danio rerio) have a third: alpha 3 chain (Colalpha3(I)). This study tests whether Colalpha3(I) is a duplicate of Colalpha1(I) by whole-genome duplication (WGD) that occurred early in the ray-fin fish lineage. We also examine how their promoter sequence was modified after WGD. We cloned Colalpha1(I), Colalpha2(I) and Colalpha3(I) cDNAs and their promoters from flounder (Paralichthys olivaceus) and obtained corresponding sequences from the genome databanks of two pufferfishes Takifugu rubripes and Tetraodon nigroviridis, by BLAST-Search using flounder sequences. Phylogenetic analysis of N-terminal sequences of ca. 100 amino acids, including signal peptide and N-propeptide sequences before short triple helical domain, indicates that Colalpha3(I), found only in teleosts, is a duplicate of Colalpha1(1) by WGD. Colalpha1(I) and Colalpha3(I) genes begin to be transcribed at different stages of Takifugu embryogenesis, suggesting that their structure of promoter is modified differently after WGD. In flounder, Takifugu and Tetraodon, the structure of proximal region of promoter is highly conserved within Colalpha1(I) and within Colalpha3(I); no homology is apparent except for the TATA element motif between Colalpha1(I) and Colalpha3(I) of each species. Unexpectedly, zebrafish Colalpha1(I) promoter is more homologous to Colalpha3(I) of flounder and fugu than Colalpha1(I) is. These results suggest that each duplicated Colalpha1(I) gene promoter inherited a unique structure after WGD, but the manner of modification differed between the phylogenetically separated zebrafish and flounder/pufferfish lineages.

17.
Vaccine ; 23(33): 4273-82, 2005 Jul 21.
Article in English | MEDLINE | ID: mdl-16005117

ABSTRACT

The function and plasticity of the developing immune system during embryonic life has been central to immunological thinking for half a century. A classical view is that antigen encountered during fetal life induces a state of acquired immunological tolerance. However, the ability to develop T cell immune responses during the perinatal period would be of great importance against intracellular pathogens. Recent experiments have challenged this notion and shown that neonatal tolerance can be circumvented by extrinsic immunological manipulations. Here, we used DNA immunization targeted at B lymphocytes to induce a CD4 T cell response that could be measured 2 weeks after birth. We conclude that T cell immunity can be programmed in utero by manipulating the parameters of the immune response in the fetal environment. Furthermore, our data suggest that under appropriate conditions the fetal immune system can be programmed to immunity.


Subject(s)
DNA/administration & dosage , Fetus/drug effects , Immune Tolerance/drug effects , Immunity/drug effects , Vaccines, Synthetic/administration & dosage , Animals , Animals, Newborn/immunology , DNA/immunology , Fetus/immunology , Immune Tolerance/immunology , Immunity/immunology , Mice , Vaccination , Vaccines, Synthetic/immunology
18.
Transplantation ; 78(9): 1274-82, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548963

ABSTRACT

BACKGROUND: Clinical application of in utero transplantation (IUT) in human fetuses with intact immune systems resulted in a very low level of donor chimerism. In this study, we examined whether the fetal immune system early in the second trimester of pregnancy (13.5 dpc) can initiate immune tolerance for major histocompatibility complex (MHC)-mismatched embryonic stem (ES) cells. We also examined whether immune tolerance mechanisms respond differently to ontogenetically different stem cells. METHODS: MHC-mismatched ES, fetal liver (FL), and bone-marrow (BM) cells (H-2kd) at 1 x 10(9) cells/kg fetal body weight were injected intraperitoneally into 13.5 dpc BALB/c fetuses (H-2Kd). Peripheral chimerism was determined in blood by flow cytometry (sensitivity< or =0.1%) at monthly intervals. Donor-specific immune responses were determined by cytotoxic lymphocyte (CTL) assay, mixed lymphocyte reaction, and T helper (Th)1 and Th2 cytokine assays. Chimeric mice at the age of 9 months received postnatal boosts (PB) with minimal conditioning of 200 cGy by intravenous injection of 1 x 10(9) of the corresponding cells/kg body weight. RESULTS: After IUT with ES, FL, or BM cells, the level of peripheral chimerism within the first 9 months of life was 0% to 0.4%. PB with 1 x 10(9)/kg of corresponding cells resulted in a decrease in the peripheral chimerism to 0% within 2 weeks of PB. CTL and cytokine assays before and after PB demonstrated a shift toward immunity. CONCLUSIONS: Immunologic tolerance was not achieved after IUT of murine fetuses at 13.5 dpc with MHC-mismatched ES cells, and only a low level chimerism was achieved.


Subject(s)
Chimerism , Fetus/immunology , Immune Tolerance , Stem Cell Transplantation , Animals , Cytokines/biosynthesis , Female , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL
19.
Biochem Biophys Res Commun ; 324(4): 1333-9, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15504360

ABSTRACT

One of the most important issues in stem cell research is to understand the regulatory mechanisms responsible for their differentiation. An extensive understanding of mechanism underlying the process of differentiation is crucial in order to prompt stem cells to perform a particular function after differentiation. To elucidate the molecular mechanisms responsible for the hematopoietic differentiation of embryonic stem cells (ESCs), we investigated murine ES cells for the presence of hematopoietic lineage markers as well as Wnt signaling pathway during treatments with different cytokines alone or in combination with another. Here we report that Wnt/beta-catenin signaling is down-regulated in hematopoietic differentiation of murine ES cells. We also found that differentiation induced by the interleukin-3, interleukin-6, and erythropoietin combinations resulted in high expression of CD3e, CD11b, CD45R/B220, Ly-6G, and TER-119 in differentiated ES cells. A high expression of beta-catenin was observed in two undifferentiated ES cell lines. Gene and protein expression analysis revealed that the members downstream of Wnt in this signaling pathway including beta-catenin, GSK-3beta, Axin, and TCF4 were significantly down-regulated as ES cells differentiated into hematopoietic progenitors. Our results show that the Wnt/beta-catenin signaling pathway plays a role in the hematopoietic differentiation of murine ESCs and also may support beta-catenin as a crucial factor in the maintenance of ES cells in their undifferentiated state.


Subject(s)
Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Cell Differentiation , Cell Line , Cytoskeletal Proteins/genetics , Down-Regulation , Mice , Signal Transduction , Trans-Activators/genetics , Wnt Proteins , beta Catenin
20.
Biotechniques ; 37(1): 81-3, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15283204

ABSTRACT

The phage is used as a scaffold to display recombinant libraries of peptides, which provides the means to rescue and amplify peptides that bind target macromolecules. Many reports showed that the T7 phage display method can be used to obtain a ligand-binding peptidefor tissue-targeted therapies in adult animals. In utero tissue targeting of fetal tissues may help in the correction of many genetic and metabolic diseases. Here we demonstrate the distribution and detection of T7 phage displaying the C-X7-C peptide library in mouse fetal tissues after systemic injection of T7 phage into pregnant mouse tail vein. T7 phage was recovered from fetal tissues 15 min after injection of T7 phage. Our results suggest that T7 phage may be a useful tool in selecting the tissue-specific ligand-binding peptide for fetal tissues. This approach may be helpful in designing in utero tissue-targeted therapies.


Subject(s)
Bacteriophage T7 , Uterus/virology , Animals , Bacteriophage T7/metabolism , Female , Gene Transfer Techniques , Immunohistochemistry , Mice , Peptides/analysis , Pregnancy , Viral Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...