Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2008 Jan 20;1(1):2–21.

Stem Cells, Phenotypic Inversion, and Differentiation

Robert W Siggins II 2,3, Ping Zhang 1,2,3, David Welsh 1, Nicole J LeCapitaine 2,3, Steve Nelson 1,2,3
PMCID: PMC2596332  PMID: 19079683

Abstract

Stem cells possess the potential to cure a myriad of ailments ranging from congenital diseases to illnesses acquired through the physiological process of aging. In the adult, these cells are extremely rare and often difficult to isolate in numbers sufficient to apply to medical treatment. Ex vivo expansion of these cells will be required for most meaningful interventions. The discovery of stem/progenitor cell inversion offers a new avenue for obtaining sufficient numbers of stem cells. Adult progenitor cells are much more common than quiescent stem cells and can be isolated with minimal interventions; therefore, inversion of progenitors to stem cells may become a feasible approach for therapeutic purposes. Stem cells are known to possess few mitochondria, and mitochondrial biogenesis is required for stem cell differentiation. The microtubule cytoskeleton is a major regulator for mitochondrial biogenesis. Investigations in the area of controlling cell differentiation and inducing phenotypic inversion, possibly through manipulation of mitochondrial biogenesis, may contribute to stem cell-based therapies.

Keywords: Stem cells, inversion, differentiation, mitochondria, microtubules

Introduction

Stem cells possess a great potential to transdifferentiate [1-3]. Bone marrow stem cells have been shown to form bone, skeletal muscle, cardiac muscle, endothelium, liver and lung parenchyma, and neural tissue [4-10]. Recently, stem cells have emerged as a promising therapy for a variety of disorders, including neurodegenerative diseases, spinal injuries, and infarcted myocardium [4, 11, 12]. At the present time, however, knowledge about the fundamental properties of stem cells, including understanding their differentiation potential and plasticity, as well as how to direct stem cells to differentiate to a desired lineage, remains limited. This review will discuss the evolution of stem cell research with emphasis on the concept of a stem cell continuum, phenotypic inversion, and influence of mitochondrial and microtubule properties on stem cell differentiation.

Historical Overview

Virchow first proposed an undifferentiated cell population in the human body in 1855. Shortly thereafter, Julius Conheim expanded Virchow's concept while engaged in research to determine the origin of cancer [13,14]. Conheim proposed that a vestigial pool of cells, which he termed “embryonic rest,” remained intact from the developing fetus into adulthood. While this idea alludes to stem cells, the origin of our modern concept of a primitive pool of cells regulating tissue function would not become realized for another 50 years. In 1917, Pappenheim speculated that an undifferentiated cell gave rise to progenitor and precursor cells of all the blood lineages [15]. This theory guided research until 1961 when McCulloch and Till published their definitive study describing colony-forming units (CFUs) in the mouse spleen after irradiation and whole bone marrow transplantation [16, 17]. In their studies, McCulloch and Till put forward two important caveats to define stem cells: the ability of self-renewal and multipotentiality. Without a thorough understanding of the surface phenotype, stem cells could only be identified certainly by their ability to form colonies in the spleens of irradiated, recipient mice. Nevertheless, this was the first report to unequivocally identify hematopoietic stem cells (HSCs).

Studies in stem cell biology remained focused on hematopoiesis in this period until Gail Martin discovered embryonic stem cells (ESCs) in 1981 [18]. This report described cells isolated from preimplantation mice, that when injected into adult mice, could produce teratocarcinomas. This property is indicative of the pluripotent nature of ESCs. Human ESCs were discovered 17 years later, when James Thomson isolated a cell derived from the early blastocyst that had high telomerase activity and lacked phenotypic markers of any known cell lineages [19]. In 1990, Potten and Loeffler refined the definition of stem cells as: “undifferentiated cells capable of (a) proliferation, (b) self maintenance, (c) the production of a large number of differentiated, functional progeny, (d) regenerating the tissue after injury, and (e) a flexibility in the use of these options [20].” The one component missing from Potten's and Loeffler's definition is the concept that stem cells must reside in niches. In 1978, Schofield first theorized the niche concept as the cellular environment which retains the stem cell and prevents its further maturation [21]. In 2000, Xie and Spradling proved the histological existence and the function of the niche for germ line stem cells in the ovary of Drosophila melanogaster [22]. Three years later, the niches for hematopoietic stem cells were also identified [23, 24].

Endogenous adult stem cells (ASCs) from tissues other than the bone marrow have been identified since 1990. ASCs were discovered first by Potten and Loeffler in the intestinal crypt [20]. A major breakthrough occurred in 1999, with two independent reports describing a resident pool of ASCs in the brain, previously thought to be a post-mitotic organ [25, 26]. In 2001, a multipotential cell was discovered in the dermis of adult mice that was capable of differentiating into neurons, glia, smooth muscle cells, and adipocytes [27]. In 2003, the existence of a multipotential progenitor cell was observed in the mouse heart, another organ thought to be post-mitotic [28]. Lately, a previously overlooked cell from the bone marrow was described as being a very small embryonic-like stem cell (VSEL-SC) that displayed characteristics of totipotent, zygote cells [29]. This discovery might confirm Virchow's and Conheim's original concept of the embryonic rest. These studies, performed in a variety of tissues representing all germ layers and containing resident stem cell pools, have created a new dogma that all adult tissues likely possess a regulatory stem cell population for homeostatic turnover and tissue maintenance.

With the development of investigation in the fields of ESCs and ASCs, methodology for isolating stem cells was rapidly evolving. Of particular interest, the use of vital dyes emerged as a tool for isolating ASCs into short-and long-term repopulating HSCs (STHSCs and LTHSCs). Rhodamine-123 (Rho-123) in 1985 and Hoechst 33342 in 1996 were used to isolate side population cells [30, 31]. LTHSCs were originally speculated to efflux dyes through the multidrug resistance (MDR) transporters. However, in 1998 Spangrude showed that low fluorescence observed in LTHSC populations stained with Rho-123 was indicative of low mitochondrial activity, and not dye efflux [32]. This elegant study set a precedent for identifying stem cells by examining the mitochondrial activity of these cells.

ESCs are considered pluripotent (capable of differentiating into all germ layers), while ASCs are postulated by many as being only multipotent (capable of differentiating into a limited number of lineages) [33, 34]. Transdifferentiation is the ability of a cell to differentiate into an entirely different cell lineage, regardless of the original state of lineage commitment. In 1974, Selman and Kafatos reported that cuticle cells in the larval silk gland of the silkmoth were capable of “metamorphosing” into salt-producing cells without dividing [35]. The transdifferentiated cell possessed two distinct mitochondrial networks, whereas the progenitor cell contained fewer mitochondria than the differentiated cell. In 1986, Eguchi proposed a mechanism of dedifferentiation, followed by redifferentiation to explain the conversion of vertebrate pigmented epithelium to lens cells in their studies [36]. In 1998, HSC plasticity was described by Ferrari and colleagues, who showed that bone marrow cells could differentiate into muscle fibers and regenerate damaged tissue [37]. While Ferrari's results expanded the understanding of HSC multipotentiality, pluripotent properties have since been discovered in HSCs. HSCs have been shown to give rise to neural and connective tissue [38]. Research, as highlighted in these examples, is beginning to alter our thinking on the nature of hierarchical organization in stem cell commitment.

Surface Markers, Phenotypic Inversion, and the Stem Cell Continuum

A variety of surface antigens are used to isolate stem cells. These include stem cell antigen-1 (Sca-1), ckit (stem cell factor receptor), and CD34. Sca-1 was discovered 20 years ago and is specific for mouse SCs, while CD34 was first reportedly used to isolate human HSCs in 1984 [39, 40]. ckit was discovered in 1986 and was shown to be expressed on both human and murine HSCs in 1989 [41, 42]. Isolation of cells based on these methods almost always requires depletion of cells committed to a specific lineage. In terms of HSCs, the surface antigen profile is dictated by the organism studied. Mouse HSCs are lineage negative (Lin-), ckit positive (ckit+), and Sca-1 positive (Sca-1+); whereas in humans, HSCs have been shown to be Lin- and CD34 positive (CD34+) [43, 44]. In contrast, mouse HSCs are CD34 negative (CD34-) [45]. This difference between organisms is surprising, but more interesting is the surface antigen expression within a particular organism's pool of HSCs and hematopoietic progenitor cells (HPCs). The specificity of surface phenotype suggests that the stem cell is a distinct functional unit.

Mouse HSCs lose Sca-1 expression as they begin to differentiate, but ckit remains highly expressed [46]. Analysis of the two populations shows that Lin-Sca-1+ckit+ (KSL) cells form three times as many colonies as Lin-Sca-1-ckit+ cells in an in vitro CFU assay [43]. Analysis of HSCs from Sca-1 knock-out (KO) mice shows an impaired ability of these cells to repopulate the bone marrow of irradiated mice [47]. The cells isolated from these KO animals also have diminished multipotentiality. Compared to cells from wild-type (WT) animals, KO cells produced less in vitro granulocyte, erythroid, macrophage, and megakaryocyte CFU (GEMM-CFU). Weissman reported a Lin-Sca-1+ckit- population isolated from the bone marrow that was incapable of responding to hematopoietic growth factors [48]. The cells in this study did not form colonies in irradiated mouse spleens, nor did they reconstitute the bone marrow. They concluded that this population of cells is either a resting stem cell, or an entirely new subset of HSCs. The first conclusion is supported by Norio Suzuki, who found cells bound in the niche express Sca-1 and GATA2 (via GATA2-directed GFP expression) [49]. The cells in these studies were all in the G0/G1 phase of the cell cycle. Weisman's second conclusion has been supported more recently with the discovery of the aforementioned VSEL-SC, although Ratajczak's group did not analyze ckit expression on their cells [29]. Another study using the identical Lin-Sca-1+ckit- phenotype found these cells had the ability to differentiate into hepatic-like cells in the absence of cell fusion [50].

A phenomenon recently described by Quesenberry's laboratory contradicts the notion of a hierarchy between stem and progenitor cells [51]. They isolated stem cells on the basis of Hoechst 33342 efflux and mitochondrial potential, rather than surface phenotype. The benefit to this method is that all the isolated cells are quiescent and/or synchronized in the cell cycle, which allowed them to correlate the phenomenon to cell cycle stage. His group used in vitro assays to analyze high-proliferative potential colony forming cells (HPP-CFC) and colony-forming unit cultures (CFU-c). In vivo competitive repopulation assays were used in conjunction to the HPP-CFC and CFU-c assays to determine HSC versus HPC. The results showed an inverse relationship between HPP-CFCs and engraftment ability. The cell cycle status was also analyzed, and cells in the S/G2 phase of the cell cycle where found to have a decreased ability to engraft when compared to cells in G0/G1. The results confirmed that this defect of engraftment was reversible with cell cycle transit. Quesenberry termed this phenomenon a progenitor/stem cell inversion.

When Quesenberry examined the surface phenotype ex vivo of cells isolated from BrdU treated mice, he found that 22% of the KSL, Thy1.1lo cells were in S-phase [52]. The Weissman laboratory sorted cells that were Lin-ckit+Sca-1+Thy1.1lo and then resorted for cells in G0, G1, or S-G2/M [53]. They then tested engraftment capacity in limited dilution competitive reconstitution assays. Only cells in G0 were able to rescue lethally irradiated mice for >12 weeks. This conclusion argues strongly for cell cycle dependence. Additionally, Jocobsen showed that KSL cells lose their self-renewal capacity when they become positive for flt-3 [54]. In fact, KSL, flt3+ cells show an increased differentiation potential towards the lymphoid lineage, and only serve as STHSCs for the myeloid lineage. KSL cells are considered stem cells, but without further depletion of HPC markers, many cells in these studies might represent lineage-committed cells. In a similar study, Ogawa's laboratory isolated KSL cells and then looked at CD34 and CD38 expression in murine HSCs [55]. They discovered a reciprocal relationship between CD34 and CD38. In vitro, cytokine stimulation shifted expression towards the CD34+ phenotype, and in vivo reconstitution experiments with KSL, CD34+ cells, showed that when the recipient bone marrow achieved steady-state, the cells were predominantly KSL, CD34-, CD38+ in mice (Figure 1). This study highlights the fact that surface phenotype of the HSC population should be considered heterogeneous and that progenitors have the potential for inversions.

Figure 1.

Figure 1

Murine Stem/Progenitor Cell Inversion in the Niche. (1) CD38+ stem cell attached to stromal cell in niche. (2) Stem cell undergoing asymmetric division. (3) CD38+ stem cell remains in the niche. (4) CD34+ progenitor cell receives signals from the microenvironment. (5) CD34+ progenitor cell may leave the niche and continue differentiating, or invert and remain in the niche as a new CD38+ stem cell.

Bernstein examined Sca-1 in C2C12 myoblastic cells in vitro [56]. These cells contain a side population subset that resembles stem cells. The results show that Sca-1 is transiently upregulated upon cell cycle withdrawal when C2C12 cells are induced to form myotubes. When Sca-1 mRNA expression was inhibited with either antisense nucleotides or protein with monoclonal antibodies, the C2C12 cells remained proliferative and did not form myotubes. If this characteristic of Sca-1 expression is the same in stem cells, then in order to amplify the stem cell pool, Sca-1 would be expected to be down regulated. As aforementioned in stem cells, Sca-1 appears to be the most primitive marker [43]. Together with Quesenberry's, Weissman's, and Bernstein's results, Sca-1 might be transiently down regulated in stem cells in a cell cycle-dependent manner [51, 54, 56]. In our recent studies, we have observed that Lin-ckit+Sca-1- progenitor cells can invert to a KSL stem cell phenotype both in vitro and in vivo in response to a variety of inflammatory stimulants (unpublished data).

Stem cells have been shown to dedifferentiate in other model systems, which also suggest a continuum exists. Spradling's group showed that cystocytes in both larvae and adult Drosophila can revert to stem-like cells [57]. In the larvae stage, these cells differentiated after depletion of the stem cell pool with cytotoxic agents or heat shock. In adult flies, overexpression of decapentaplegic (dpp), the Drosophila homologue to the known stem cell regulator bone morphogenic protein 4 (BMP-4), was able to stimulate a reversion to a more primitive germline stem cell phenotype.

The expression patterns of HPCs and HSCs are remarkably similar; therefore inversions should not be surprising. Catherine Laiosa, Matthias Stadtfeld, and Thomas Graf reviewed this aspect extensively, and showed that a single transcription factor (TF) can convert HPCs of one lineage into a separate lineage; however, the cells in these studies were not synchronized in the cell cycle [58]. Altering either PU1.1 or GATA1 expression can transdifferentiate the myeloid and lymphoid precursors. Quesenberry reported hotspots at defined points in the cell cycle of TF expression [59]. Early in S-phase, cells showed a reversible propensity to differentiate to the megakaryocyte lineage. While late in S-phase, the cells were predisposed to the granulocyte lineage—also reversibly. Quesenberry's group also showed that TFs of the megakaryocyte lineage were expressed during the differentiation hotspot.

Recent investigations have reported the creation of embryonic-like stem cells from fully differentiated cells. Two groups, Marius Wernig and Kazutosh Takahashi and Shinya Yamanaka reprogrammed mouse fibroblasts with ectopic expression of four transcription factors: POU domain, class 5, transcription factor (Oct3/4), sex determining region Y-box2 (Sox2), cellular myelocytomatosis oncogene (cMyc), and Kruppel-like factor 4 (Klf4) [60, 61]. In 2007, human fibroblasts were also reprogrammed to an ESC-like state, termed induced pluripotent stem (iPS) cells, by transfection with the previously mentioned TFs or with Sox2, Oct4, NANOG, and Lin28 [62,63]. These TFs are all highly enriched in pluripotent stem cells. Further, Wernig analyzed chromatin remodeling and found epigenetic markers similar to ESCs. This would suggest that genomic alterations are important for the dedifferentiated phenotype. Nevertheless, the cytosolic milieu appears to be more important in determination of stem cell phenotype. Cloning studies have been instrumental in furthering this hypothesis. Nuclear transfer of adult, differentiated nuclei into enucleated ova creates totipotent stem cells as seen in the early zygote. Wilmut and Campbell produced live lambs from adult mammary gland, fetus, and embryo to confirm that adult cells are capable of forming stem cells [64]. These results showed definitively that genetic modifications are not the sole determinant for a differentiated phenotype.

Transfer of adult nuclei into oocytes and the subsequent generation of stem cells suggests that other mechanisms, besides chromatin and TF expression, are responsible for stem cell plasticity. Isolation of HSCs based on cell surface antigen expression invariably gives a heterogeneous population of HSCs and HPCs. Using mitochondrial energetics, with Rho-123 fluorescence, and Hoechst 33342 efflux isolates a population known to be quiescent. Intriguingly, the same percentage of HSCs is recovered when cells are sorted for low Rho-123 fluorescence or Sca-1 positivity [65]. Either group is equally capable of hematopoietic reconstitution. Other markers of energy metabolism have also been used in the isolation of HSCs and stem cells in general. Daniel Pearce examined human cord blood surface phenotype and showed a strong correlation with aldehyde dehydrogenase (AHD) activity [66]. Greater than 90% of CD34+CD38- cells and CD34+CD133+ cells were also positive for high AHD activity. These two surface phenotypes are considered HSC populations. Michael Kastan and John Hilton compared AHD activity between HPCs and lymphocytes, and found high AHD activity in HPCs [67]. High activity levels of AHD protect the stem cells from retinaldehyde and oxidative stress during the transition from anaerobic glycolysis to oxidative phosphorylation (OXPHOS) for energy production [68]. This, with the data discussed earlier regarding Sca-1 as the most primitive marker, should lead research to more closely examine energy metabolism in Sca-1+ and KSL cells.

Mitochondrial Properties and Stem Cells

Mitochondria are as heterogeneous as the tissues in which they are examined. Each cell type possesses a certain range of mitochondria per cell [69]. Proteomes vary dramatically between cell types, and mitochondrial DNA (mtDNA) copy number may be one to ten per organelle. Fundamental properties, such as subcellular localization, mitochondrial membrane potential (ΔΨm), and reticular networks are also different between cell types [70, 71]. These facts suggest that a particular cell type might be identified based on these mitochondrial parameters.

Jankovic examined the role of Id1, a helix-loop-helix transcription factor, in myeloid differentiation [72]. Using Id1 KO mice, they discovered that HSCs prematurely entered into myeloid commitment, and also inappropriately expressed myeloerythroid genes. Tanaka and Finkel also examined this inhibitor of differentiation and found that Id1 was able to produce apoptosis in neonatal and adult cardiomyocytes, but not in endothelial cells (ECs) or fibroblasts [73]. In seeking to determine the mechanism of action, they discovered that Id1 functions through a redox-dependent mechanism. Cardiomyocytes are almost entirely dependent on mitochondria for ATP production, whereas ECs and fibroblasts rely more heavily on glycolysis [75]. These results offer clues as to the mechanisms of differentiation as they pertain to energy metabolism.

The environments of the uterus before placentation and the marrow surrounding HSC niches are anaerobic [75]. Feasibly, this prevents oxidative damage to the early zygote. To produce ATP in this environment, stem cells, from the fertilized oocytes to the adult stem cells, rely heavily on glycolysis for cellular ATP production, and thus do not require a large number of mitochondria [76]. As such, any lineage commitment requires an increase in OXPHOS and, subsequently, an increase in mitochondrial ATP production. This is achieved through two processes: mitochondrial differentiation and mitochondrial biogenesis [77]. Mitochondrial differentiation is simply the activation of mitochondria. This is most often equated with an increase in the ΔΨm. Mitochondrial activation correlates to many processes like subcellular localization, but Ca++ is probably the main effector [78, 79]. In fact, isolation of stem cells based on Rho-123 is indicative of undifferentiated mitochondria. Biogenesis involves replication of mitochondrial genomes, as well as an increase in all proteins necessary to form a new mitochondrion from an existing organelle.

Mitochondrial biogenesis occurs through the process of binary fission, similar to their bacterial progenitors [80]. The regulation of this process is tissue specific, but all tissues examined require the expression of the peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), beta (PGC-1β), or PPAR-γ, coactivator-related 1 (PPRC) [81-83]. This protein family regulates many of the transcription factors for both mitochondrial-encoded genes and nuclear-encoded genes, in particular mitochondrial transcription factor A (tFAM) and nuclear respiratory factor 1 and 2 (NRF-1 and NRF-2) [82]. Studies have shown that mitochondrial biogenesis is particularly important to stem cell differentiation [84-88]. This complex process is known to encompass about 20% of total cellular protein [89].

The peroxisome proliferators-activated receptor (PPAR) family of TFs has been shown to be crucial in the control of cellular metabolism, by regulating genes of the different metabolic pathways [90, 91]. PGC family members differentially recruit the PPAR TFs under different conditions. PPAR-alpha (PPAR-α) has been shown to be crucially important in fatty acid metabolism, while PPAR-gamma (PPAR-γ) has been extensively studied in gluconeogenesis [92]. PPAR-γ, in conjunction with PGC-1α increases mitochondrial biogenesis [93]. PPAR-γ has also been shown to inhibit canonical wingless homologue (Wnt) signaling, which is a crucial pathway in maintaining an undifferentiated state [94]. Notch signaling has been shown to upregulate PPAR-γ [95]. Notch has been studied mostly in its ability to direct differentiation towards specific lineages [96-98]. Wnt and Notch signaling antagonize each other, and this might be due to influences of PPAR-γ [99].

The many molecular pathways governing mitochondrial biogenesis have varying importance depending on the tissue. In skeletal muscle and granulosa cells, calcium (Ca++) is a major initiator of biogenesis [100, 101]. In adipose tissue, nitric oxide (NO), thyroid hormone, and rosiglitazones have been the most intensively analyzed pathways for increased mitochondrial biogenesis [102-104]. NO and hydrogen peroxide (H2O2) increase biogenesis in B cell chronic lymphocytic leukemia (CLL) and osteosarcoma cells, respectively [105, 106]. All of these initiators, either directly or indirectly, lead to an increased expression of PGC-1α [107-110].

In vivo PGC-1α is essential for the increased expression of nuclear-derived transcription factors involved in mitochondrial biogenesis, which are primarily mitochondrial transcription factor A (TFAM) and nuclear respiratory factor 1 (NRF1) [82]. PPRC performs similar functions as PGC-1α, but this coactivator is not regulated by thermogenesis [111]. However, PPRC is expressed under in vitro conditions if PGC-1α is not. These two regulatory proteins have complementary functions.

The activation of PGC-1α expression in vivo is under the direct control of both the myocyte enhancer factor 2 (MEF2) and the forkhead box O (FOXO) families of transcription factors [112, 113]. In addition, cyclic adenosine monophosphate (cAMP) response element-binding (CREB) proteins interact with cAMP response elements (CRE) in the promoter of PGC-1α to activate transcription [114]. v-akt murine thymoma viral oncogene homolog (AKT) phosphorylation of FOXO sequesters the TF to the cytoplasm, leading to FOXO inactivation; however, PKA phosphorylation of MEF2 increases transcriptional activity [115, 116]. Members of the p38 mitogen-activated protein kinase (MAPK) family interact with AKT and are involved primarily in cellular responses to environmental stress [117]. p38 MAPK phosphorylates MEF2 family members. p38 increases both the transcription of AKT and the activation of AKT through phosphorylation [118, 119]. Additionally, p38 MAPK phosphorylates PGC-1α protein, increasing its nuclear localization [120].

PGC-1α can interact with both MEF2 and FOXO family members to increase its own expression in a feed-forward loop [119]. In skeletal muscle, Ca++ acts as a potent activator of MEF2 transcription through calcium/calmodulin-dependent protein kinase IV (CaMKIV) and calcineurin. CaMKIV increases CREB activity through phosphorylation and activates calcineurin [121]. Calcineurin dephosphorylates nuclear factor of activated T-cells (NFAT), which translocates to the nucleus where it interacts with MEF2 to activate PGC-1α expression [122]. NFAT knock-out (KO) mice die at embryonic day 10.5 and present with diminished ventricular myocyte proliferation containing abnormal, swollen mitochondria [123]. NFAT transcription and translation are enhanced by p38 MAPK, but phosphorylation of NFAT by p38 sequesters it in the cytoplasm [124]. Activated calcineurin leads to p38 inactivation [125]. In ESCs, bone morphogenic protein 4 (BMP4) inhibits p38 MAPK allowing for SC self-renewal [126]. Conversely, reactive oxygen species (ROS) upregulate p38, leading to increased HSC turnover and SC exhaustion [127]. p38 and Ca++ initiate the signals involved in PGC-1α regulation, thereby increasing mitochondrial biogenesis and causing differentiation in a level and time-dependent manner (Figure 2).

Figure 2.

Figure 2

Signal Transduction Pathway for Regulators of PGC-1α Expression and Mitochondrial Biogenesis. A) Regulators of PGC-1α are interconnected in a complex network of positive and negative feedback loops. This ensures that a prolonged signal favoring mitochondrial biogenesis is required for enlargement of the mitochondrial pool. B) Transcriptional control of PGC-1α expression and mitochondrial biogenesis.

Mitochondrial mass and activity increases with differentiation [128-131]. Inhibition of mitochondrial biogenesis prevents terminal differentiation [82, 132]. This suggests cells possessing a progenitor phenotype will maintain a progenitor phenotype without increases in mitochondrial mass. Similarly, mitosis in a progenitor without a concomitant increase in mitochondrial number could feasibly lead to the production of stem cells. Increased mitochondrial mass is seen in germline stem cells, ESCs, and adult stem cells when they are induced to differentiate [129, 131, 133].

The literature is replete with methods for the in vitro differentiation of stem cells. Three popular agents that have been used to induce stem cell differentiation in vitro are dexamethasone (DEX), retinoic acid (RA), and dimethyl sulfoxide (DMSO) [134-141]. DEX is a glucocorticoid receptor (GR) agonist, and RA activates the retinoic acid receptor (RAR/RXR) family. These agents have been shown to upregulate transcripts for both PGC-1α and PGC-1β, and RAR/RXR forms complexes with PGC-1α [142, 143]. DMSO activates mitochondrial Ca++ signaling, affecting differentiation through a different mechanism [141, 144]. DEX has been shown to play a crucial role in mitochondrial biogenesis in a tissue-specific manner [145]. In adrenal-ectomized rats treated with DEX, liver mitochondrial content is returned to sham-operated control values, but heart mitochondrial mass was unchanged. DEX directly increases PPAR-γ expression.

When PPAR-γ is induced in mouse bone marrow (BM) or peripheral blood, angiogenic progenitor cells differentiated towards the endothelial progenitor cell (EPC) lineage [146]. These cells concomitantly showed a decreased potential to differentiate into the smooth muscle cell lineage. PPAR-γ induction also increases the multipotential murine cell line, C3H10T1/2, to differentiate towards the adipocytes lineage, while inhibiting osteoblast differentiation [128]. Conversely, treatment with RA led to differentiation into the osteogenic lineage [147]. Increased mitochondrial mass accompanied the different-tiation in these cells. In human bone marrow cells, DEX enhanced osteoblast differentiation [137]. In a separate study, mouse mesenchymal stem cells (MSCs) preferentially differentiated towards the adipocytes lineage, and were inhibited from the osteogenic lineage [138]. These results appear to be conflicting; however, the intracellular milieu and species of origin may determine the end effect of DEX treatment.

The selective differentiation of DEX is also seen in monomyelocytic HPCs [135]. DEX rapidly caused neutriphil differentiation, whereas vitamin D3 led to monocyte/macrophage lineage commitment. It is important to note that PGC-1α has been shown to coactivate the D3 receptor [143]. Different blood cell lineages display preferential expression of PPAR family members [148]. Myeloid cells predominantly express PPAR-γ, while lymphocytes mostly express PPAR-α. Intriguingly, DEX treatment of erythroid progenitors actually leads to proliferation, but RA treatment causes differentiation [136]. These results suggest that a finely tuned mechanism controlling the metabolic state of stem cells is active in determining different-tiation capacity.

RA is the active metabolite formed from retinaldehyde, a downstream product from dietary retinol [149]. PGC-1α is a coactivator for the RAR/RXR family of nuclear receptors [143]. As aforementioned, PGC-1α plays a major role in mitochondrial biogenesis. Aldehyde dehydrogenase (AHD) is a key enzyme in the production of RA [149]. Three AHD isoforms are largely responsible: AHD2, AHD7, and AHD-M1 [150]. The first two are cytoplasmic and the last one is mitochondrial. Stem cells and progenitor cells have been shown to express high levels of AHD activity, and this fact has been used in their isolation [66].

Similar to DEX treatment, RA displays differential effects on differentiation in a time-and tissue-dependent manner. This is likely due to the large number of different RAR/RXR receptors. Treatment of murine ESCs with RA during the first two days of development promoted neurogenesis, but inhibited cardiomyogenesis [144]. Treatment for two to five days increased skeletal muscle and adipocyte differentiation, in addition to neurogenesis. Treatment with RA after five days promoted cardiomyogenesis and smooth muscle cell differentiation, while inhibiting the myogenic and adipogenic lineages. Human ESCs could not be induced to differentiate into cardiomyocytes with RA, displaying species differences or ESC heterogeneity in RA's mechanism of action. In promyeloid progenitor cells, RA has been shown to drive commitment to the granulocyte lineage [151]. However, in KSL stem cells, RA appears to maintain the quiescent, undifferentiated state [152]. RA has also been shown to alter the lineage of differentiated cells. When white adipocytes are treated with RA, these cells display gene expression profiles and properties of brown adipose tissue (BAT) [153]. BAT is enriched for mitochondria as compared to white adipose tissue (WAT) [154]. Similar increases in mitochondria are seen in murine ESCs. RA consistently increased the level of transcripts for tFAM and polymerase G, the major polymerase for mtDNA replication [86].

ΔΨm, representing the mitochondrial proton-motive force, is responsible for controlling Ca++ uptake, respiratory rate, mitochondrial ATP production, and reactive oxygen species (ROS) production [130, 155, 156]. Mitochondria within a single cell have varying ΔΨm, depending on the subcellular localization and the relative health of the mitochondrion [70]. Most often, researchers consider ΔΨm by averaging intensity for a single cell or multiple cells due to the prohibitive nature of studying single mitochondrion in situ. Analyzing the average ΔΨm offers an adequate appraisal of total cellular oxidative metabolism.

Rho-123 is commonly employed to analyze ΔΨm, since incorporation of the fluorophore increases with increasing ΔΨm [32]. As previously mentioned, LTHSCs can be isolated from the bone marrow using Rho-123. Morphologic analysis of Rho-123low cells revealed small mitochondria with diffuse cytoplasmic distribution, whereas Rho-123high cells had two-fold larger mitochondria and a perinuclear distribution. The modest increase in mitochondrion size would not account for the thirty-fold increase observed in Rho-123 fluorescence. Additionally, cell cycle analysis showed that 3% of Rho-123low and 33% of Rho-123high cells were in the S/G2 phase. These data suggest that cell cycle status alone is a poor indicator of engraftability. In accordance with this data, osteoprogenitor cells (OPCs) induced to differentiate show an increased retention of Rho-123 in a time- and concentration-dependent manner [157]. Two peaks in Rho-123 are seen at days 4 and 10 in both control and induced cultures, with the induced cultures retaining more dye at all time points. This fluctuation seen in the uninduced OPCs is similar to the cyclic changes observed in ΔΨm from single, isolated neural mitochondria [158].

In single mitochondrion analysis, the oscillations in ΔΨm from total depolarization to physiological levels occurred at a rate of one per minute, and they were shown to be Ca++- dependent. In murine erythroleukemia cells (MEL) induced to differentiate with DMSO, ΔΨm remains relatively constant for the first 10 hours of treatment [141]. A total collapse in ΔΨm occurs prior to the appearance of differentiated progeny. This decay in potential corresponds to a release of mitochondrial Ca++ stores. Depolarizing the mitochondria with an uncoupling agent before induction eliminated the 10 hour lag time for MEL differentiation. Again, this depolarization corresponded to increased cytosolic Ca++ concentration. In P19 cells, DMSO treatment almost instantly causes an increase in cytoplasmic Ca++ [144]. These cells express a very early gene of the mesoderm lineage, Brachyury T. DMSO also causes a similar Ca++ rise in mouse embryonal carcinoma cells, which may lead to their differentiation. This is consistent with the role mitochondria fulfill as intracellular Ca++ buffers, representing a possible feedback mechanism for PGC-1α regulation. Thus, ΔΨm controls differentiation by enhancing mitochondrial biogenesis through Ca++ signaling and by increasing ATP supply through OXPHOS.

Since ΔΨm is responsible for the rate of ATP synthesis, cytosolic ATP levels would be expected to regulate membrane potential. In fact, the Ca++-dependent fluctuations seen in ΔΨm from isolated mitochondria are stabilized by micromolar concentrations of ATP and ADP [158]. The mammalian target of rapamycin (mTOR), a downstream kinase in the AKT pathway and chemotherapy target, has recently been described as a cytosolic ATP sensor, independent of its ability to sense amino acid (AA) concentrations [159]. The classical functions of mTOR have been studied in terms of regulating ribosomal biogenesis and cell growth, and increasing protein translation rates by phosphorylation of S6 kinase 1 (S6K1) and initiation factor 4E binding protein 1 (4E-BP1). mTOR and mTOR/raptor complexes have recently been purified from mitochondrial fractions [160]. This colocalization with the mitochondria was abolished by treatment with rapamycin. Additionally, rapamycin treatment and small interfering RNA (siRNA) directed towards mTOR binding partners lead to a decrease in ΔΨm, oxygen consumption, and ATP production. Thus, mTOR integrates signals from nutrient concentration, ATP levels, and growth factor stimulation in order to modify the translational response of the cell, and functions in a feed-forward manner to increase mitochondrial activity.

Perhaps the best evidence to date suggesting a role for mitochondrial biogenesis, ΔΨm, and subcellular localization in affecting different-tiation is in the study of oocytes [161, 162]. Mitochondria are few in number and low in activity level in immature oocytes. The subcellular distribution is diffuse. As the oocytes mature, activity level increases, and distribution becomes pericytoplasmic. Mitochondrial ΔΨm becomes intermediate up until the 16-cell blastocyst stage, after which it decreases. The distribution of mitochondria in inner mass cells, from which ESCs arise, is perinuclear, and the ΔΨm remains low. Cultured ESCs retain these parameters, and loss of this subcellular localization is correlated to differentiation or quiescence [163].

Microtubules in the Regulation of Mitochondria and Stem Cell Differentiation

Microtubules (MTs) are the most important protein in the control of organelle subcellular localization, including the mitochondria [164]. This function is critical during interphase, but during the G2/M phase of the cell cycle, an additional microtubule (MT) network forms called the mitotic spindle [165]. The mitotic spindle is regulated by the MT organizing center (MTOC). The MTOC is comprised of a centriole, which forms the centrosome. During mitosis, the location of the maternal centriole is responsible for determining the cleavage plane of the stem cell [166, 167].

Redistribution of the MT network corresponds to differentiation. MT distribution changes during development [168]. As the fertilized oocytes proceed through the 8-cell stage, MTs reorganize from perilamellar to a perinuclear arrangement. This correlates to the mitochondrial distribution early in development [161]. The centriole, as expected, also reorganizes in a similar manner. As epithelial cells mature, the typical non-polarized progenitor cell acquires an apico-basal organization [169]. Centrioles migrate to the nascent apical surface, and reorganize the MT cytoskeleton from a radial to an apico-basal distribution.

Maternal centrioles reproduce once during the course of the cell cycle [170]. After replication, the daughter centriole migrates to the opposite end of the cell from the maternal centriole. The mature maternal centriole is capable of forming a more stabilized MT network [171]. This is largely due to a larger number of MT associated proteins (MAPs) remaining with the maternal centriole. In adult germline stem cells in Drosophila, the stem cell is anchored to the supporting cells of the niche [170]. When these cells undergo asymmetric division to form a daughter progenitor cell, the stem cell retains the mature, original centriole. Two hypotheses exist for this phenomenon: the more organized MT network maintains the mitochondrial localization and prevents biogenesis, or the maternal centriole acts a basal body, creating a primary cilium that serves as a nurse cell communication domain [172]. A primary cilium has a large surface area-to-volume ratio, and could feasibly act as an amplifying center for any signals sent by the nurse cell to maintain quiescence.

Regardless of the actual mechanism, this phenomenon of asymmetric division, where the stem cell retains the proximity of the niche, is seen in many tissues of multiple organisms. In skeletal muscle of mice, satellite cells are paired box gene 7 positive (Pax7+) and myogenic factor 5 negative (Myf5+) [173]. As these cells undergo asymmetric division, the basal cell retains the satellite cell phenotype, while the apical cell becomes Myf5 positive (Myf5+). When these two populations were transplanted into injured muscle, Pax7+Myf5+ cells contribute to the satellite cell pool, and the Pax7+Myf5+ cells undergo marked differentiation into new muscle cells.

Niche proximity and mitotic spindle orientation also plays a role in final progenitor cell commitment. In the developing mouse brain, cleavage plane determines the fate of the neural progenitor cell (NPC) [174]. While a cleavage plane parallel to the niche did not inhibit differentiation in all cases, a perpendicular cleavage plane always resulted in a symmetric division that produced two neurons. These studies taken together suggest that mitotic spindle stability is a function of the maternal centriole, and that disparities in centriole maturity can determine symmetric versus asymmetric division and commitment of progeny.

Mitochondrial biogenesis can be altered by MT stability. The mechanisms involved are not clear, but Wnt signaling and cytoplasmic Ca++ concentration might be the modulator of this process. Wnt signaling increases MT stability through activation of janus kinase (JNK) and inhibition of glycogen synthase kinase 3 beta (Gsk3β). As mentioned above, Wnt signaling is downregulated by PPAR-γ [175]. This suggests a link between MT stability and major TFs involved in mitochondrial biogenesis. The adapter protein, myeloid differentiation primary response protein-5 (MyD88-5), has been demonstrated to link MTs and mitochondria and to coordinate JNK localization to the mitochondria [176]. When MTs are destabilized, MyD88-5 dissociates from tubulin. In cells with high MyD88-5 expression, mitochondria localize around the MTOC. MT organization is required for optimal store operated calcium release (SOCE) from the endoplasmic reticulum (ER), through regulating the location of the ubiquitously expressed Ca++ channel activator, STIM1 [177]. The perinuclear arrangement of mitochondria in stem cells, therefore could allow for proper cytoplasmic-mitochondrial-ER Ca++ homeostasis. As stated previously, Ca++ has an essential role in mitochondrial biogenesis.

MT stability also appears to be regulated by p38 MAPK. Disruption of p38 attenuates MT disruption by tumor necrosis factor alpha (TNFα) and nocodazole, a pharmacological MT disrupter [178, 179]. Environmental stress, such as oxidative damage, activates p38; however the role of p38 in oxidant-induced MT disruption have not been examined [180]. Nevertheless, H2O2 and NO (from iNOS) cause MT disruption, and both of these oxidants lead to increased mitochondrial biogenesis [181, 182]. Interestingly, MT stability does appear to have opposing effects depending on the state of differentiation. When human keritinocytes (skin SCs) are treated with a MT disrupting agent, E-cadherin-catenin complexes are reorganized to the cell periphery [183]. These complexes anchor the SCs to the niche cells, maintaining quiescence. When the same treatment is performed in progenitor cells, no relocalization of this junctional-signalling complex is observed. These data allude to the fact that mitochondrial biogenesis is required for final commitment, and before biogenesis occurs, progenitor cells may invert to stem cells.

When MTs are destabilized by heterozygous deletion of survivin, a MAP, in mouse HPCs, decreased formation of enucleated erythroid cells is seen [184]. In human CD34+ HPCs, the peptide, shepherdin, causes survivin depletion. At low levels of shepherdin treatment, HPCs show diminished erythroid burst-forming units. At higher shepherdin concentrations, granulocyte-macrophage CFUs (GM-CFUs) and granulocyte erythrocyte macrophage megakaryocyte CFUs (GEMMCFUs) are also inhibited. This research, with the above centriole localization studies suggest a mechanism by which differentiation hotspots and inversions might correlate to cell cycle status. However, this cell cycle-dependency of differentiation potential might only relate to the maturity of the MT network and the level of mitochondrial biogenesis.

Perspectives

Understanding stem cell differentiation is critically important for the clinical application of these cells. At the present time, expansion of stem cells ex vivo is required for conducting potential treatments. Controlling the differentiation state of stem cells will increase the efficacy of these treatments. In addition, seemingly contradictory results from clinical trials with stem cells might be eliminated as we acquire a better understanding of their phenotypic variations, particularly the role of the mitochondria and their biophysical properties in regulating phenotypic changes [185, 186]. The phenomenon of stem cell inversions could represent a new mechanism in the regulation of adult stem cell turnover. Further studies on this mechanism may provide novel information for understanding how stem cells maintain tissue homeostasis. Advancement of knowledge in this area will form a foundation upon which new strategies of stem cell therapy will be developed. The progenitor pool of most stem cell compartments is considered rapidly dividing and greatly outnumbers the stem cell pool. Therefore isolation of progenitors could yield a sufficient number of inverted stem cells in order to support many new therapeutic protocols with minimal interventions in patients.

Acknowledgments

This work was supported by NIH grants AA09803, HL075161, HL073770, and HL76100.

References

  • 1.Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair–current views. Stem Cells. 2007;25:2896–902. doi: 10.1634/stemcells.2007-0637. [DOI] [PubMed] [Google Scholar]
  • 2.Hayashi Y, Tsuji S, Tsujii M, Nishida T, Ishii S, Nakamura T, Eguchi H, Kawano S. The transdifferentiation of bone-marrow-derived cells in colonic mucosal regeneration after dextran-sulfate-sodium-induced colitis in mice. Pharmacology. 2007;80:193–9. doi: 10.1159/000104148. [DOI] [PubMed] [Google Scholar]
  • 3.Udani VM. The continuum of stem cell transdifferentiation: possibility of hematopoietic stem cell plasticity with concurrent CD45 expression. Stem Cells Dev. 2006;15:1–3. doi: 10.1089/scd.2006.15.1. [DOI] [PubMed] [Google Scholar]
  • 4.Ikeda T. 2007. Stem cells and neonatal brain injury. Cell Tissue Res (In Press)
  • 5.Song SJ, Jeon O, Yang HS, Han DK, Kim BS. Effects of culture conditions on osteogenic differentiation in human mesenchymal stem cells. J Microbiol Biotechnol. 2007;17:1113–9. [PubMed] [Google Scholar]
  • 6.Abedi M, Foster BM, Wood KD, Colvin GA, McLean SD, Johnson KW, Greer DA. Haematopoietic stem cells participate in muscle regeneration. Br J Haematol. 2007;138:792–801. doi: 10.1111/j.1365-2141.2007.06720.x. [DOI] [PubMed] [Google Scholar]
  • 7.Pasha Z, Wang Y, Sheikh R, Zhang D, Zhao T, Ashraf M. 2007. Preconditioning enhances cell survival and differentiation of stem cells during transplantation in infarcted myocardium. Cardiovasc Res (In Press) [DOI] [PubMed]
  • 8.Ohnishi S, Ohgushi H, Kitamura S, Nagaya N. Mesenchymal stem cells for the treatment of heart failure. Int J Hematol. 2007;86:17–21. doi: 10.1532/IJH97.07041. [DOI] [PubMed] [Google Scholar]
  • 9.Qihao Z, Xigu C, Guanghui C, Weiwei Z. Spheroid formation and differentiation into hepatocyte-like cells of rat mesenchymal stem cell induced by co-culture with liver cells. DNA Cell Biol. 2007;26:497–503. doi: 10.1089/dna.2006.0562. [DOI] [PubMed] [Google Scholar]
  • 10.Sueblinvong V, Loi R, Eisenhauer PL, Bernstein IM, Suratt BT, Spees JL, Weiss DJ. 2007. Derivation of lung epithelium from human cord blood-derived mesenchymal stem cells. Am J Respir Crit Care Med (In Press) [DOI] [PMC free article] [PubMed]
  • 11.Mitsui T, Shumsky JS, Lepore AC, Murray M, Fischer I. Transplantation of neuronal and glial restricted precursors into contused spinal cord improves bladder and motor functions, decreases thermal hypersensitivity, and modifies intraspinal circuitry. J Neurosci. 2005;25:9624–36. doi: 10.1523/JNEUROSCI.2175-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Caplice NM. The future of cell therapy for acute myocardial infarction. Nat Clin Pract Cardiovasc Med. 2006;3:S129–32. doi: 10.1038/ncpcardio0432. [DOI] [PubMed] [Google Scholar]
  • 13.Virchow R. Editorial Archive fuer pathologische. Anatomie und Physiologie fuer klinische Medizin. 1855;8:23–54. [Google Scholar]
  • 14.Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J, Kucia M. A hypothesis for an embryonic origin of pluripotent Oct-4þ stem cells in adult bone marrow and other tissues. Leukemia. 2007;21:860–7. doi: 10.1038/sj.leu.2404630. [DOI] [PubMed] [Google Scholar]
  • 15.Pappenheim A. Prinzipien der neueren morphologischen Haematozytologie nach zytogenetischer Grundlage. Folia Haematologica. 1917;21:91. [Google Scholar]
  • 16.Till JE, McCulloch EA. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiation Research. 1961;14:213–22. [PubMed] [Google Scholar]
  • 17.Simlnovitch L, McCulloch EA, Till JE. The distribution of colony-forming cells among spleen colonies. J Cell Physiol. 1963;62:327–36. doi: 10.1002/jcp.1030620313. [DOI] [PubMed] [Google Scholar]
  • 18.Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA. 1981;78:7634–8. doi: 10.1073/pnas.78.12.7634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–7. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  • 20.Potten CS, Loeffler M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development. 1990;110:1001–20. doi: 10.1242/dev.110.4.1001. [DOI] [PubMed] [Google Scholar]
  • 21.Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell: A hypothesis. Blood Cells. 1978;4:7–25. [PubMed] [Google Scholar]
  • 22.Xie T, Spradling AC. A niche maintaining germ line stem cells in the Drosophila ovary. Science. 2000;290:328–30. doi: 10.1126/science.290.5490.328. [DOI] [PubMed] [Google Scholar]
  • 23.Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S, Wiedemann LM, Mishina Y, Li L. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836–41. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
  • 24.Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–6. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 25.Doetsch F, Caillé I, Lim DA, García-Verdugo JM, Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 1999;97:703–16. doi: 10.1016/s0092-8674(00)80783-7. [DOI] [PubMed] [Google Scholar]
  • 26.Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Frisen J. Identification of a neural stem cell in the adult mammalian central nervous system. Cell. 1999;96:25–34. doi: 10.1016/s0092-8674(00)80956-3. [DOI] [PubMed] [Google Scholar]
  • 27.Toma JG, Akhavan M, Fernandes KJL, Barnabé-Heider F, Sadikot A, Kaplan DR, Miller FD. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nature Cell Biology. 2001;3:778–84. doi: 10.1038/ncb0901-778. [DOI] [PubMed] [Google Scholar]
  • 28.Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
  • 29.Zuba-Surma EK, Kucia M, Abdel-Latif A, Dawn B, Hall B, Singh R, Lillard JW, Ratajczak MZ. Morphological characterization of very small embryonic- like stem cells (VSELs) by image stream system analysis. J Cell Mol Med. 2007;20:1499–514. doi: 10.1111/j.1582-4934.2007.00154.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bertoncello I, Hodgson GS, Bradley TR. Multiparameter analysis of transplantable hemopoietic stem cells: I. The separation and enrichment of stem cells homing to marrow and spleen on the basis of rhodamine-123 fluorescence. Exp Hematol. 1985;13:999–1006. [PubMed] [Google Scholar]
  • 31.Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 1996;183:1797–806. doi: 10.1084/jem.183.4.1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kim M, Cooper DD, Hayes SF, Spangrude GJ. Rhodamine-123 staining in hematopoietic stem cells of young mice indicates mitochondrial activation rather than dye efflux. Blood. 1998;91:4106–17. [PubMed] [Google Scholar]
  • 33.Wulf GG, Jackson KA, Goodell MA. Somatic stem cell plasticity: Current evidence and emerging concepts. Experimental Hematology. 2001;29:1361–70. doi: 10.1016/s0301-472x(01)00752-4. [DOI] [PubMed] [Google Scholar]
  • 34.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, MoSca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 35.Selman K, Kafatos FC. Transdifferentiation in the labial gland of silk moths: Is DNA required for cellular metamorphosis? Cell Differentiation. 1974;3:81–94. doi: 10.1016/0045-6039(74)90030-x. [DOI] [PubMed] [Google Scholar]
  • 36.Itoh Y, Eguchi G. In vitro analysis of cellular metaplasia from pigmented epithelial cells to lens phenotypes: A unique model system for studying cellular and molecular mechanisms of “transdifferentiation”. Developmental Biology. 1986;115:353–362. doi: 10.1016/0012-1606(86)90255-1. [DOI] [PubMed] [Google Scholar]
  • 37.Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, Mavilio F. Muscle regeneration by bone marrow–derived myogenic progenitors. Science. 1998;279:1528–30. doi: 10.1126/science.279.5356.1528. [DOI] [PubMed] [Google Scholar]
  • 38.Sigurjonsson OE, Perreault MC, Egeland T, Glover JC. Adult human hematopoietic stem cells produce neurons efficiently in the regenerating chicken embryo spinal cord. Proc Natl Acad Sci USA. 2005;102:5227–32. doi: 10.1073/pnas.0501029102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yutoku M, Grossberg AL, Pressman D. A cell surface antigenic determinant present on mouse plasmacytes and only about half of mouse thymocytes. J Immunol. 1974;112:1774–81. [PubMed] [Google Scholar]
  • 40.Civin CI, Strauss LC, Brovall C, Fackler MJ, Schwartz JF, Shaper JH. Antigenic Analysis of Hematopoiesis III. A Hematopoietic Progenitor Cell Surface Antigen Defined by a Monoclonal Antibody Raised against KG-la Cells. J Immunol. 1984;133:157–65. [PubMed] [Google Scholar]
  • 41.Besmer P, Murphy JE, George PC, Qiu F, Bergold PJ, Lederman L, Snyder HW, Brodeur D, Zuckerman EE, Hardy WD. A new acute transforming feline retrovirus and relationship of its oncogene v-kit with the protein kinase gene family. Nature. 1986;320:415–21. doi: 10.1038/320415a0. [DOI] [PubMed] [Google Scholar]
  • 42.André C, d'Auriol L, Lacombe C, Gisselbrecht S, Galibert F. c-kit mRNA expression in human and murine hematopoietic cell lines. Oncogene. 1989;4:1047–9. [PubMed] [Google Scholar]
  • 43.Okada S, Nakauchi H, Nagayoshi K, Nishikawa S, Miura Y, Suda T. In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells. Blood. 1992;80:3044–50. [PubMed] [Google Scholar]
  • 44.Novelli EM, Ramirez M, Civin CI. Biology of CD34+ CD38- cells in lymphohematopoiesis. Leuk Lymphoma. 1998;31:285–93. doi: 10.3109/10428199809059221. [DOI] [PubMed] [Google Scholar]
  • 45.Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 1996;273:242–5. doi: 10.1126/science.273.5272.242. [DOI] [PubMed] [Google Scholar]
  • 46.Katayama N, Shih J, Nishikawa S, Kina T, Clark SC, Ogawa M. Stage-specific expression of c-kit protein by murine hematopoietic progenitors. Blood. 1993;82:2353–60. [PubMed] [Google Scholar]
  • 47.Ito CY, Li CYJ, Bernstein A, Dick JE, Stanford WL. Hematopoietic stem cell and progenitor defects in Sca-1/Ly-6A–null mice. Blood. 2003;101:517–23. doi: 10.1182/blood-2002-06-1918. [DOI] [PubMed] [Google Scholar]
  • 48.Randall TD, Weissman IL. Characterization of a population of cells in the bone marrow that phenotypically mimics hematopoietic stem cells: Resting stem cells or mystery population? Stem Cells. 1998;16:38–48. doi: 10.1002/stem.160038. [DOI] [PubMed] [Google Scholar]
  • 49.Suzuki N, Ohneda O, Minegishi N, Nishikawa M, Ohta T, Takahashi S, Engel JD, Yamamoto M. Combinatorial Gata2 and Sca1 expression defines hematopoietic stem cells in the bone marrow niche. Proc Natl Acad Sci USA. 2006;103:2202–7. doi: 10.1073/pnas.0508928103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yamada Y, Nishimoto E, Mitsuya H, Yonemura Y. In vitro transdifferentiation of adult bone marrow Sca-1+ cKit- cells cocultured with fetal liver cells into hepatic-like cells without fusion. Exp Hematol. 2006;34:97–106. doi: 10.1016/j.exphem.2005.09.018. [DOI] [PubMed] [Google Scholar]
  • 51.Colvin GA, Lambert JF, Moore BE, Carlson JE, Dooner MS, Abedi M, Cerny J, Quesenberry PJ. Intrinsic hematopoietic stem cell/progenitor plasticity: Inversions. J Cell Physiol. 2004;199:20–31. doi: 10.1002/jcp.10436. [DOI] [PubMed] [Google Scholar]
  • 52.Quesenberry PJ, Colvin G, Dooner G, Dooner M, Aliotta JM, Johnson K. The stem cell continuum: cell cycle, injury, and phenotype lability. Ann N Y Acad Sci. 2007;1106:20–9. doi: 10.1196/annals.1392.016. [DOI] [PubMed] [Google Scholar]
  • 53.Passegué E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med. 2005;202:1599–611. doi: 10.1084/jem.20050967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Adolfsson J, Borge OJ, Bryder D, Theilgaard-Mönch K, Astrand-Grundström I, Sitnicka E, Sasaki Y, Jacobsen SE. Upregulation of Flt3 expression within the bone marrow Lin(-)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity. Immunity. 2001;15:659–69. doi: 10.1016/s1074-7613(01)00220-5. [DOI] [PubMed] [Google Scholar]
  • 55.Tajima F, Deguchi T, Laver JH, Zeng H, Ogawa M. Reciprocal expression of CD38 and CD34 by adult murine hematopoietic stem cells. Blood. 2001;97:2618–24. doi: 10.1182/blood.v97.9.2618. [DOI] [PubMed] [Google Scholar]
  • 56.Epting CL, López JE, Shen X, Liu L, Bristow J, Bernstein HS. Stem cell antigen-1 is necessary for cell-cycle withdrawal and myoblast differentiation in C2C12 cells. J Cell Sci. 2004;117:6185–95. doi: 10.1242/jcs.01548. [DOI] [PubMed] [Google Scholar]
  • 57.Fuller MT, Spradling AC. Male and female Drosophila germline stem cells: two versions of immortality. Science. 2007;316:402–4. doi: 10.1126/science.1140861. [DOI] [PubMed] [Google Scholar]
  • 58.Laiosa CV, Stadtfeld M, Graf T. Determinants of lymphoid-myeloid lineage diversification. Annu Rev Immunol. 2006;24:705–38. doi: 10.1146/annurev.immunol.24.021605.090742. [DOI] [PubMed] [Google Scholar]
  • 59.Colvin GA, Dooner MS, Dooner GJ, Sanchez-Guijo FM, Demers DA, Abedi M, Ramanathan M, Chung S, Pascual S, Quesenberry PJ. Stem cell continuum: directed differentiation hotspots. Exp Hematol. 2007;35:96–107. doi: 10.1016/j.exphem.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 60.Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, Bernstein BE, Jaenisch R. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature. 2007;448:318–24. doi: 10.1038/nature05944. [DOI] [PubMed] [Google Scholar]
  • 61.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 62.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 63.Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
  • 64.Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997;385:810–3. doi: 10.1038/385810a0. [DOI] [PubMed] [Google Scholar]
  • 65.Ivanova NB, Dimos JT, Schaniel C, Hackney JA, Moore KA, Lemischka IR. A stem cell molecular signature. Science. 2002;298:601–4. doi: 10.1126/science.1073823. [DOI] [PubMed] [Google Scholar]
  • 66.Pearce DJ, Taussig D, Simpson C, Allen K, Rohatiner AZ, Lister TA, Bonnet D. Characterization of cells with a high aldehyde dehydrogenase activity from cord blood and acute myeloid leukemia samples. Stem Cells. 2005;23:752–60. doi: 10.1634/stemcells.2004-0292. [DOI] [PubMed] [Google Scholar]
  • 67.Kastan MB, Schlaffer E, Russo JE, Colvin OM, Civin CI, Hilton J. Direct demonstration of elevated aldehyde dehydrogenase in human hematopoietic progenitor cells. Blood. 1990;75:1947–50. [PubMed] [Google Scholar]
  • 68.Tsuji H, Meguro N, Suzuki Y, Tsutsumi N, Hirai A, Nakazono M. Induction of mitochondrial aldehyde dehydrogenase by submergence facilitates oxidation of acetaldehyde during re-aeration in rice. FEBS Lett. 2003;546:369–73. doi: 10.1016/s0014-5793(03)00631-8. [DOI] [PubMed] [Google Scholar]
  • 69.St John JC, Amaral A, Bowles E, Oliveira JF, Lloyd R, Freitas M, Gray HL, Navara CS, Oliveira G, Schatten GP, Spikings E, RamalhoSantos J. The analysis of mitochondria and mitochondrial DNA in human embryonic stem cells. Methods Mol Biol. 2006;331:347–74. doi: 10.1385/1-59745-046-4:347. [DOI] [PubMed] [Google Scholar]
  • 70.Nesti C, Pasquali L, Vaglini F, Siciliano G, Murri L. The role of mitochondria in stem cell biology. Biosci Rep. 2007;27:165–71. doi: 10.1007/s10540-007-9044-1. [DOI] [PubMed] [Google Scholar]
  • 71.Twig G, Graf SA, Wikstrom JD, Mohamed H, Haigh SE, Elorza A, Deutsch M, Zurgil N, Reynolds N, Shirihai OS. Tagging and tracking individual networks within a complex mitochondrial web with photoactivatable GFP. Am J Physiol Cell Physiol. 2006;291:C176–84. doi: 10.1152/ajpcell.00348.2005. [DOI] [PubMed] [Google Scholar]
  • 72.Jankovic V, Ciarrocchi A, Boccuni P, De Blasio T, Benezra R, Nimer SD. Id1 restrains myeloid commitment, maintaining the self-renewal capacity of hematopoietic stem cells. Proc Natl Acad Sci USA. 2007;104:1260–5. doi: 10.1073/pnas.0607894104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tanaka K, Pracyk JB, Takeda K, Yu ZX, Ferrans VJ, Deshpande SS, Ozaki M, Hwang PM, Lowenstein CJ, Irani K, Finkel T. Expression of Id1 results in apoptosis of cardiac myocytes through a redox-dependent mechanism. J Biol Chem. 1998;273:25922–8. doi: 10.1074/jbc.273.40.25922. [DOI] [PubMed] [Google Scholar]
  • 74.Shah AM, Mebazaa A, Yang ZK, Cuda G, Lankford EB, Pepper CB, Sollott SJ, Sellers JR, Robotham JL, Lakatta EG. Inhibition of myocardial crossbridge cycling by hypoxic endothelial cells: a potential mechanism for matching oxygen supply and demand? Circ Res. 1997;80:688–98. doi: 10.1161/01.res.80.5.688. [DOI] [PubMed] [Google Scholar]
  • 75.Izquierdo JM, Luis AM, Cuezva JM. Postnatal mitochondrial differentiation in rat liver. Regulation by thyroid hormones of the beta-subunit of the mitochondrial F1-ATPase complex. J Biol Chem. 1990;265:9090–7. [PubMed] [Google Scholar]
  • 76.Thompson JE, Conlon JP, Yang X, Sanchez PV, Carroll M. Enhanced growth of myelodysplastic colonies in hypoxic conditions. Exp Hematol. 2007;35:21–31. doi: 10.1016/j.exphem.2006.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Izquierdo JM, Ricart J, Ostronoff LK, Egea G, Cuezva JM. Changing patterns of transcriptional and post-transcriptional control of beta-F1-ATPase gene expression during mitochondrial biogenesis in liver. J Biol Chem. 1995;270:10342–50. doi: 10.1074/jbc.270.17.10342. [DOI] [PubMed] [Google Scholar]
  • 78.Simpson PB, Russell JT. Mitochondrial Ca2+ uptake and release influence metabotropic and ionotropic cytosolic Ca2+ responses in rat oligodendrocyte progenitors. J Physiol. 1998;508:413–26. doi: 10.1111/j.1469-7793.1998.413bq.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Vergun O, Reynolds IJ. Fluctuations in mitochondrial membrane potential in single isolated brain mitochondria: modulation by adenine nucleotides and Ca2+ Biophys J. 2004;87:3585–93. doi: 10.1529/biophysj.104.042671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kuroiwa T. The discovery of the division apparatus of plastids and mitochondria. J Electron Microsc. 2000;49:123–34. doi: 10.1093/oxfordjournals.jmicro.a023776. [DOI] [PubMed] [Google Scholar]
  • 81.Soriano FX, Liesa M, Bach D, Chan DC, Palacín M, Zorzano A. Evidence for a mitochondrial regulatory pathway defined by peroxisome proliferator-activated receptor-gamma coactivator-1 alpha, estrogen-related receptor-alpha, and mitofusin 2. Diabetes. 2006;55:1783–91. doi: 10.2337/db05-0509. [DOI] [PubMed] [Google Scholar]
  • 82.Gleyzer N, Vercauteren K, Scarpulla RC. Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators. Mol Cell Biol. 2005;25:1354–66. doi: 10.1128/MCB.25.4.1354-1366.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lelliott CJ, Medina-Gomez G, Petrovic N, Kis A, Feldmann HM, Bjursell M, Parker N, Curtis K, Campbell M, Hu P, Zhang D, Litwin SE, Zaha VG, Fountain KT, Boudina S, Jimenez-Linan M, Blount M, Lopez M, Meirhaeghe A, Bohlooly M, Storlien L, Stromstedt M, Snaith M, Oresnic M, Abel ED, Cannon B, Vidal-Puig A. Ablation of PGC-1b results in defective mitochondrial activity, thermogenesis, hepatic function, and cardiac performance. PLoS Biology. 2006;4:2042–2056. doi: 10.1371/journal.pbio.0040369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wilson-Fritch L, Burkart A, Bell G, Mendelson K, Leszyk J, Nicoloro S, Czech M, Corvera S. Mitochondrial biogenesis and remodeling during adipogenesis and in response to the insulin sensitizer rosiglitazone. Mol Cell Biol. 2003;23:1085–94. doi: 10.1128/MCB.23.3.1085-1094.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Funes JM, Quintero M, Henderson S, Martinez D, Qureshi U, Westwood C, Clements MO, Bourboulia D, Pedley RB, Moncada S, Boshoff C. Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc Natl Acad Sci USA. 2007;104:6223–8. doi: 10.1073/pnas.0700690104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Facucho-Oliveira JM, Alderson J, Spikings EC, Egginton S, St John JC. Mitochondrial DNA replication during differentiation of murine embryonic stem cells. J Cell Sci. 2007;120:4025–34. doi: 10.1242/jcs.016972. [DOI] [PubMed] [Google Scholar]
  • 87.St John JC, Ramalho-Santos J, Gray HL, Petrosko P, Rawe VY, Navara CS, Simerly CR, Schatten GP. The expression of mitochondrial DNA transcription factors during early cardiomyocyte in vitro differentiation from human embryonic stem cells. Cloning Stem Cells. 2005;7:141–53. doi: 10.1089/clo.2005.7.141. [DOI] [PubMed] [Google Scholar]
  • 88.Chung S, Dzeja PP, Faustino RS, Perez-Terzic C, Behfar A, Terzic A. Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells. Nat Clin Pract Cardiovasc Med. 2007;4:S60–7. doi: 10.1038/ncpcardio0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Goffart S, Wiesner RJ. Regulation and coordination of nuclear gene expression during mitochondrial biogenesis. Exp Physiol. 2003;88:33–40. doi: 10.1113/eph8802500. [DOI] [PubMed] [Google Scholar]
  • 90.Dello Russo C, Gavrilyuk V, Weinberg G, Almeida A, Bolanos JP, Palmer J, Pelligrino D, Galea E, Feinstein DL. Peroxisome proliferator-activated receptor gamma thiazolidinedione agonists increase glucose metabolism in astrocytes. J Biol Chem. 2003;278:5828–36. doi: 10.1074/jbc.M208132200. [DOI] [PubMed] [Google Scholar]
  • 91.Guzmán M, Lo Verme J, Fu J, Oveisi F, Blázquez C, Piomelli D. Oleoylethanolamide stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-alpha) J Biol Chem. 2004;279:27849–54. doi: 10.1074/jbc.M404087200. [DOI] [PubMed] [Google Scholar]
  • 92.Herzig S, Hedrick S, Morantte I, Koo SH, Galimi F, Montminy M. CREB controls hepatic lipid metabolism through nuclear hormone receptor PPAR-gamma. Nature. 2003;426:190–3. doi: 10.1038/nature02110. [DOI] [PubMed] [Google Scholar]
  • 93.Bogacka I, Xie H, Bray GA, Smith SR. Pioglitazone induces mitochondrial biogenesis in human subcutaneous adipose tissue in vivo. Diabetes. 2005;54:1392–9. doi: 10.2337/diabetes.54.5.1392. [DOI] [PubMed] [Google Scholar]
  • 94.Katoh M, Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer Res. 2007;13:4042–5. doi: 10.1158/1078-0432.CCR-06-2316. [DOI] [PubMed] [Google Scholar]
  • 95.Garcés C, Ruiz-Hidalgo MJ, Font de Mora J, Park C, Miele L, Goldstein J, Bonvini E, Porrás A, Laborda J. Notch-1 controls the expression of fatty acid-activated transcription factors and is required for adipogenesis. J Biol Chem. 1997;272:29729–34. doi: 10.1074/jbc.272.47.29729. [DOI] [PubMed] [Google Scholar]
  • 96.Yamamoto N, Tanigaki K, Han H, Hiai H, Honjo T. Notch/RBP-J signaling regulates epidermis/hair fate determination of hair follicular stem cells. Curr Biol. 2003;13:333–8. doi: 10.1016/s0960-9822(03)00081-2. [DOI] [PubMed] [Google Scholar]
  • 97.Wakamatsu Y, Maynard TM, Weston JA. Fate determination of neural crest cells by NOTCH-mediated lateral inhibition and asymmetrical cell division during gangliogenesis. Development. 2000;127:2811–21. doi: 10.1242/dev.127.13.2811. [DOI] [PubMed] [Google Scholar]
  • 98.Milner LA, Bigas A. Notch as a mediator of cell fate determination in hematopoiesis: Evidence and speculation. Blood. 1999;93:2431–48. [PubMed] [Google Scholar]
  • 99.Foltz DR, Santiago MC, Berechid BE, Nye JS. Glycogen synthase kinase-3beta modulates notch signaling and stability. Curr Biol. 2002;12:1006–11. doi: 10.1016/s0960-9822(02)00888-6. [DOI] [PubMed] [Google Scholar]
  • 100.Wu H, Kanatous SB, Thurmond FA, Gallardo T, Isotani E, Bassel-Duby R, Williams RS. Regulation of mitochondrial biogenesis in skeletal muscle by CaMK. Science. 2002;296:349–52. doi: 10.1126/science.1071163. [DOI] [PubMed] [Google Scholar]
  • 101.Yeh TS, Ho JD, Yang VW, Tzeng CR, Hsieh RH. Calcium stimulates mitochondrial biogenesis in human granulosa cells. Ann N Y Acad Sci. 2005;1042:157–62. doi: 10.1196/annals.1338.017. [DOI] [PubMed] [Google Scholar]
  • 102.Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L, Cantoni O, Clementi E, Moncada S, Carruba MO, Nisoli E. TNF-alpha downregulates eNOS expression and mitochondrial biogenesis in fat and muscle of obese rodents. J Clin Invest. 2006;116:2791–8. doi: 10.1172/JCI28570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Weitzel JM, Iwen KA, Seitz HJ. Regulation of mitochondrial biogenesis by thyroid hormone. Experimental Physiology. 2003;88:121–8. doi: 10.1113/eph8802506. [DOI] [PubMed] [Google Scholar]
  • 104.Rong JX, Qiu Y, Hansen MK, Zhu L, Zhang V, Xie M, Okamoto Y, Mattie MD, Higashiyama H, Asano S, Strum JC, Ryan TE. Adipose mitochondrial biogenesis is suppressed in db/db and high-fat diet-fed mice and improved by rosiglitazone. Diabetes. 2007;56:1751–60. doi: 10.2337/db06-1135. [DOI] [PubMed] [Google Scholar]
  • 105.Carew JS, Nawrocki ST, Xu RH, Dunner K, McConkey DJ, Wierda WG, Keating MJ, Huang P. Increased mitochondrial biogenesis in primary leukemia cells: the role of endogenous nitric oxide and impact on sensitivity to fludarabine. Leukemia. 2004;18:1934–40. doi: 10.1038/sj.leu.2403545. [DOI] [PubMed] [Google Scholar]
  • 106.Lee CF, Liu CY, Hsieh RH, Wei YH. Oxidative stress-induced depolymerization of microtubules and alteration of mitochondrial mass in human cells. Ann N Y Acad Sci. 2005;1042:246–54. doi: 10.1196/annals.1338.027. [DOI] [PubMed] [Google Scholar]
  • 107.Borniquel S, Valle I, Cadenas S, Lamas S, Monsalve M. Nitric oxide regulates mitochondrial oxidative stress protection via the transcriptional coactivator PGC-1alpha. FASEB J. 2006;20:1889–91. doi: 10.1096/fj.05-5189fje. [DOI] [PubMed] [Google Scholar]
  • 108.McClure TD, Young ME, Taegtmeyer H, Ning XH, Buroker NE, López-Guisa J, Portman MA. Thyroid hormone interacts with PPARalpha and PGC-1 during mitochondrial maturation in sheep heart. Am J Physiol Heart Circ Physiol. 2005;289:H2258–64. doi: 10.1152/ajpheart.00473.2005. [DOI] [PubMed] [Google Scholar]
  • 109.Hondares E, Mora O, Yubero P, Rodriguez de la Concepción M, Iglesias R, Giralt M, Villarroya F. Thiazolidinediones and rexinoids induce peroxisome proliferator-activated receptor-coactivator (PGC)-1alpha gene transcription: an autoregulatory loop controls PGC-1alpha expression in adipocytes via peroxisome proliferator-activated receptor-gamma coactivation. Endocrinology. 2006;147:2829–38. doi: 10.1210/en.2006-0070. [DOI] [PubMed] [Google Scholar]
  • 110.Finkel T. Cell biology: A clean energy programme. Nature. 2006;444:151–2. doi: 10.1038/444151a. [DOI] [PubMed] [Google Scholar]
  • 111.Scarpulla RC. Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene. 2002;286:81–9. doi: 10.1016/s0378-1119(01)00809-5. [DOI] [PubMed] [Google Scholar]
  • 112.Czubryt MP, McAnally J, Fishman GI, Olson EN. Regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha) and mitochondrial function by MEF2 and HDAC5. Proc Natl Acad Sci USA. 2003;100:1711–6. doi: 10.1073/pnas.0337639100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Daitoku H, Yamagata K, Matsuzaki H, Hatta M, Fukamizu A. Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR. Diabetes. 2003;52:642–9. doi: 10.2337/diabetes.52.3.642. [DOI] [PubMed] [Google Scholar]
  • 114.Akimoto T, Pohnert SC, Li P, Zhang M, Gumbs C, Rosenberg PB, Williams RS, Yan Z. Exercise stimulates PGC-1alpha transcription in skeletal muscle through activation of the p38 MAPK pathway. J Biol Chem. 2005;280:19587–93. doi: 10.1074/jbc.M408862200. [DOI] [PubMed] [Google Scholar]
  • 115.Fabre S, Lang V, Harriague J, Jobart A, Unterman TG, Trautmann A, Bismuth G. Stable activation of phosphatidylinositol 3-kinase in the T cell immunological synapse stimulates Akt signaling to FoxO1 nuclear exclusion and cell growth control. J Immunol. 2005;174:4161–71. doi: 10.4049/jimmunol.174.7.4161. [DOI] [PubMed] [Google Scholar]
  • 116.Southgate RJ, Bruce CR, Carey AL, Steinberg GR, Walder K, Monks R, Watt MJ, Hawley JA, Birnbaum MJ, Febbraio MA. PGC-1alpha gene expression is down-regulated by Akt-mediated phosphorylation and nuclear exclusion of FoxO1 in insulin-stimulated skeletal muscle. FASEB J. 2005;19:2072–4. doi: 10.1096/fj.05-3993fje. [DOI] [PubMed] [Google Scholar]
  • 117.Wang X, Tang X, Li M, Marshall J, Mao Z. Regulation of neuroprotective activity of myocyte-enhancer factor 2 by cAMP-protein kinase A signaling pathway in neuronal survival. J Biol Chem. 2005;280:16705–13. doi: 10.1074/jbc.M501819200. [DOI] [PubMed] [Google Scholar]
  • 118.Keren A, Tamir Y, Bengal E. The p38 MAPK signaling pathway: a major regulator of skeletal muscle development. Mol Cell Endocrinol. 2006;252:224–30. doi: 10.1016/j.mce.2006.03.017. [DOI] [PubMed] [Google Scholar]
  • 119.Cabane C, Coldefy AS, Yeow K, Dérijard B. The p38 pathway regulates Akt both at the protein and transcriptional activation levels during myogenesis. Cell Signal. 2004;16:1405–15. doi: 10.1016/j.cellsig.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 120.Fan M, Rhee J, St-Pierre J, Handschin C, Puigserver P, Lin J, Jäeger S, Erdjument Bromage H, Tempst P, Spiegelman BM. Suppression of mitochondrial respiration through recruitment of p160 myb binding protein to PGC-1alpha: modulation by p38 MAPK. Genes Dev. 2004;18:278–89. doi: 10.1101/gad.1152204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sato K, Suematsu A, Nakashima T, Takemoto Kimura S, Aoki K, Morishita Y, Asahara H, Ohya K, Yamaguchi A, Takai T, Kodama T, Chatila TA, Bito H, Takayanagi H. Regulation of osteoclast differentiation and function by the CaMK-CREB pathway. Nat Med. 2006;12:1410–6. doi: 10.1038/nm1515. [DOI] [PubMed] [Google Scholar]
  • 122.Handschin C, Rhee J, Lin J, Tarr PT, Spiegelman BM. An autoregulatory loop controls peroxisome proliferator-activated receptor gamma coactivator 1alpha expression in muscle. Proc Natl Acad Sci USA. 2003;100:7111–6. doi: 10.1073/pnas.1232352100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bushdid PB, Osinska H, Waclaw RR, Molkentin JD, Yutzey KE. NFATc3 and NFATc4 are required for cardiac development and mitochondrial function. Circ Res. 2003;92:1305–13. doi: 10.1161/01.RES.0000077045.84609.9F. [DOI] [PubMed] [Google Scholar]
  • 124.Wu CC, Hsu SC, Shih HM, Lai MZ. Nuclear factor of activated T cells c is a target of p38 mitogen-activated protein kinase in T cells. Mol Cell Biol. 2003;23:6442–54. doi: 10.1128/MCB.23.18.6442-6454.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lim HW, New L, Han J, Molkentin JD. Calcineurin enhances MAPK phosphatase-1 expression and p38 MAPK inactivation in cardiac myocytes. J Biol Chem. 2001;276:15913–9. doi: 10.1074/jbc.M100452200. [DOI] [PubMed] [Google Scholar]
  • 126.Qi X, Li TG, Hao J, Hu J, Wang J, Simmons H, Miura S, Mishina Y, Zhao GQ. BMP4 supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein kinase pathways. Proc Natl Acad Sci USA. 2004;101:6027–32. doi: 10.1073/pnas.0401367101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, Ohmura M, Naka K, Hosokawa K, Ikeda Y, Suda T. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12:446–51. doi: 10.1038/nm1388. [DOI] [PubMed] [Google Scholar]
  • 128.Paulik MA, Lenhard JM. Thiazolidinediones inhibit alkaline phosphatase activity while increasing expression of uncoupling protein, deiodinase, and increasing mitochondrial mass in C3H10T1/2 cells. Cell Tissue Res. 1997;290:79–87. doi: 10.1007/s004410050910. [DOI] [PubMed] [Google Scholar]
  • 129.Cho YM, Kwon S, Pak YK, Seol HW, Choi YM, Park do J, Park KS, Lee HK. Dynamic changes in mitochondrial biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. Biochem Biophys Res Commun. 2006;348:1472–8. doi: 10.1016/j.bbrc.2006.08.020. [DOI] [PubMed] [Google Scholar]
  • 130.Moyes CD, Mathieu-Costello OA, Tsuchiya N, Filburn C, Hansford RG. Mitochondrial biogenesis during cellular differentiation. Am J Physiol. 1997;272:C1345–51. doi: 10.1152/ajpcell.1997.272.4.C1345. [DOI] [PubMed] [Google Scholar]
  • 131.Luo GF, Yu TY, Wen XH, Li Y, Yang GS. 2007. Alteration of mitochondrial oxidative capacity during porcine preadipocyte differentiation and in response to leptin. Mol Cell Biochem (In Press)
  • 132.Chung S, Dzeja PP, Faustino RS, Perez-Terzic C, Behfar A, Terzic A. Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells. Nat Clin Pract Cardiovasc Med. 2007;4:S60–7. doi: 10.1038/ncpcardio0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Bhat KM. The posterior determinant gene nanos is required for the maintenance of the adult germline stem cells during Drosophila oogenesis. Genetics. 1999;151:1479–92. doi: 10.1093/genetics/151.4.1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Matsuda N, Yokoyama K, Takeshita S, Watanabe M. Role of epidermal growth factor and its receptor in mechanical stress-induced differentiation of human periodontal ligament cells in vitro. Arch Oral Biol. 1998;43:987–97. doi: 10.1016/s0003-9969(98)00079-x. [DOI] [PubMed] [Google Scholar]
  • 135.Lucas T, Krugluger W, Samorapoompichit P, Gamperl R, Beug H, Förster O, Boltz-Nitulescu G. Self-renewal, maturation, and differentiation of the rat myelomonocytic hematopoietic stem cell. FASEB J. 1999;13:263–72. doi: 10.1096/fasebj.13.2.263. [DOI] [PubMed] [Google Scholar]
  • 136.Minegishi N, Minegishi M, Tsuchiya S, Fujie H, Nagai T, Hayashi N, Yamamoto M, Konno T. Erythropoietin-dependent induction of hemoglobin synthesis in a cytokine-dependent cell line M-TAT. J Biol Chem. 1994;269:27700–4. [PubMed] [Google Scholar]
  • 137.Beloti MM, Rosa AL. Osteoblast differentiation of human bone marrow cells under continuous and discontinuous treatment with dexamethasone. Braz Dent J. 2005;16:156–61. doi: 10.1590/s0103-64402005000200013. [DOI] [PubMed] [Google Scholar]
  • 138.Yin L, Li YB, Wang YS. Dexamethasone-induced adipogenesis in primary marrow stromal cell cultures: mechanism of steroid-induced osteonecrosis. Chin Med J. 2006;119:581–8. [PubMed] [Google Scholar]
  • 139.Chute JP, Muramoto GG, Whitesides J, Colvin M, Safi R, Chao NJ, McDonnell DP. Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci USA. 2006;103:11707–12. doi: 10.1073/pnas.0603806103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wobus AM, Kaomei G, Shan J, Wellner MC, Rohwedel J, Ji Guanju, Fleischmann B, Katus HA, Hescheler J, Franz WM. Retinoic acid accelerates embryonic stem cell-derived cardiac differentiation and enhances development of ventricular cardiomyocytes. J Mol Cell Cardiol. 1997;29:1525–39. doi: 10.1006/jmcc.1997.0433. [DOI] [PubMed] [Google Scholar]
  • 141.Levenson R, Macara IG, Smith RL, Cantley L, Housman D. Role of mitochondrial membrane potential in the regulation of murine erythroleukemia cell differentiation. Cell. 1982;28:855–63. doi: 10.1016/0092-8674(82)90064-2. [DOI] [PubMed] [Google Scholar]
  • 142.Lin J, Tarr PT, Yang R, Rhee J, Puigserver P, Newgard CB, Spiegelman BM. PGC-1beta in the regulation of hepatic glucose and energy metabolism. J Biol Chem. 2003;278:30843–8. doi: 10.1074/jbc.M303643200. [DOI] [PubMed] [Google Scholar]
  • 143.Delerive P, Wu Y, Burris TP, Chin WW, Suen CS. PGC-1 functions as a transcriptional coactivator for the retinoid X receptors. J Biol Chem. 2002;277:3913–7. doi: 10.1074/jbc.M109409200. [DOI] [PubMed] [Google Scholar]
  • 144.Lev S, Kehat I, Gepstein L. Differentiation pathways in human embryonic stem cell-derived cardiomyocytes. Ann N Y Acad Sci. 2005;1047:50–65. doi: 10.1196/annals.1341.005. [DOI] [PubMed] [Google Scholar]
  • 145.Prieur B, Bismuth J, Delaval E. Effects of adrenal steroid hormones on mitochondrial maturation during the late fetal period. Eur J Biochem. 1998;252:194–9. doi: 10.1046/j.1432-1327.1998.2520194.x. [DOI] [PubMed] [Google Scholar]
  • 146.Wang CH, Ciliberti N, Li SH, Szmitko PE, Weisel RD, Fedak PW, Al-Omran M, Cherng WJ, Li RK, Stanford WL, Verma S. Rosiglitazone facilitates angiogenic progenitor cell differentiation toward endothelial lineage: a new paradigm in glitazone pleiotropy. Circulation. 2004;109:1392–400. doi: 10.1161/01.CIR.0000123231.49594.21. [DOI] [PubMed] [Google Scholar]
  • 147.Yajima Y, Sato M, Sumida M, Kawashima S. Mechanism of adult primitive mesenchymal ST-13 preadipocyte differentiation. Endocrinology. 2003;144:2559–65. doi: 10.1210/en.2002-220894. [DOI] [PubMed] [Google Scholar]
  • 148.Jones DC, Ding X, Daynes RA. Nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is expressed in resting murine lymphocytes. The PPARalpha in T and B lymphocytes is both transactivation and transrepression competent. J Biol Chem. 2002;277:6838–45. doi: 10.1074/jbc.M106908200. [DOI] [PubMed] [Google Scholar]
  • 149.Chen M, Achkar C, Gudas LJ. Enzymatic conversion of retinaldehyde to retinoic acid by cloned murine cytosolic and mitochondrial aldehyde dehydrogenases. Mol Pharmacol. 1994;46:88–96. [PubMed] [Google Scholar]
  • 150.Lee MO, Manthey CL, Sladek NE. Identification of mouse liver aldehyde dehydrogenases that catalyze the oxidation of retinaldehyde to retinoic acid. Biochem Pharmacol. 1991;42:1279–85. doi: 10.1016/0006-2952(91)90266-8. [DOI] [PubMed] [Google Scholar]
  • 151.Shi C, Zhang X, Chen Z, Sulaiman K, Feinberg MW, Ballantyne CM, Jain MK, Simon DI. Integrin engagement regulates monocyte differentiation through the forkhead transcription factor Foxp1. J Clin Invest. 2004;114:408–18. doi: 10.1172/JCI21100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Purton LE. Roles of retinoids and retinoic Acid receptors in the regulation of hematopoietic stem cell self-renewal and differentiation. PPAR Res. 2007;2007:87934. doi: 10.1155/2007/87934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mercader J, Ribot J, Murano I, Felipe F, Cinti S, Bonet ML, Palou A. Remodeling of white adipose tissue after retinoic acid administration in mice. Endocrinology. 2006;147:5325–32. doi: 10.1210/en.2006-0760. [DOI] [PubMed] [Google Scholar]
  • 154.Unami A, Shinohara Y, Kajimoto K, Baba Y. Comparison of gene expression profiles between white and brown adipose tissues of rat by microarray analysis. Biochem Pharmacol. 2004;67:555–64. doi: 10.1016/j.bcp.2003.09.010. [DOI] [PubMed] [Google Scholar]
  • 155.Saotome M, Katoh H, Satoh H, Nagasaka S, Yoshihara S, Terada H, Hayashi H. Mitochondrial membrane potential modulates regulation of mitochondrial Ca2+ in rat ventricular myocytes. Am J Physiol Heart Circ Physiol. 2005;288:H1820–8. doi: 10.1152/ajpheart.00589.2004. [DOI] [PubMed] [Google Scholar]
  • 156.González A, Pariente JA, Salido GM. Ethanol stimulates ROS generation by mitochondria through Ca(2+) mobilization and increases GFAP content in rat hippocampal astrocytes. Brain Res. 2007;1178:28–37. doi: 10.1016/j.brainres.2007.08.040. [DOI] [PubMed] [Google Scholar]
  • 157.Klein BY, Gal I, Hartshtark Z, Segal D. Induction of osteoprogenitor cell differentiation in rat marrow stroma increases mitochondrial retention of rhodamine 123 in stromal cells. J Cell Biochem. 1993;53:190–7. doi: 10.1002/jcb.240530303. [DOI] [PubMed] [Google Scholar]
  • 158.Vergun O, Reynolds IJ. Fluctuations in mitochondrial membrane potential in single isolated brain mitochondria: modulation by adenine nucleotides and Ca2+ Biophys J. 2004;87:3585–93. doi: 10.1529/biophysj.104.042671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G. Mammalian TOR: a homeostatic ATP sensor. Science. 2001;294:1102–5. doi: 10.1126/science.1063518. [DOI] [PubMed] [Google Scholar]
  • 160.Schieke SM, Phillips D, McCoy JP, Jr, Aponte AM, Shen RF, Balaban RS, Finkel T. The mammalian target of rapamycin (mTOR) pathway regulates mitochondrial oxygen consumption and oxidative capacity. J Biol Chem. 2006;281:27643–52. doi: 10.1074/jbc.M603536200. [DOI] [PubMed] [Google Scholar]
  • 161.Tarazona AM, Rodríguez JI, Restrepo LF, Olivera-Angel M. Mitochondrial activity, distribution and segregation in bovine oocytes and in embryos produced in vitro. Reprod Domest Anim. 2006;41:5–11. doi: 10.1111/j.1439-0531.2006.00615.x. [DOI] [PubMed] [Google Scholar]
  • 162.Bavister BD, Squirrell JM. Mitochondrial distribution and function in oocytes and early embryos. Hum Reprod. 2000;15:189–98. doi: 10.1093/humrep/15.suppl_2.189. [DOI] [PubMed] [Google Scholar]
  • 163.Lonergan T, Brenner C, Bavister B. Differentiation-related changes in mitochondrial properties as indicators of stem cell competence. J Cell Physiol. 2006;208:149–53. doi: 10.1002/jcp.20641. [DOI] [PubMed] [Google Scholar]
  • 164.Chada SR, Hollenbeck PJ. Nerve growth factor signaling regulates motility and docking of axonal mitochondria. Curr Biol. 2004;14:1272–6. doi: 10.1016/j.cub.2004.07.027. [DOI] [PubMed] [Google Scholar]
  • 165.Niethammer P, Kronja I, Kandels-Lewis S, Rybina S, Bastiaens P, Karsenti E. Discrete states of a protein interaction network govern interphase and mitotic microtubule dynamics. PLoS Biol. 2007;5:e29. doi: 10.1371/journal.pbio.0050029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Yamashita YM, Mahowald AP, Perlin JR, Fuller MT. Asymmetric inheritance of mother versus daughter centrosome in stem cell division. Science. 2007;315:518–21. doi: 10.1126/science.1134910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Haydar TF, Ang E, Jr, Rakic P. Mitotic spindle rotation and mode of cell division in the developing telencephalon. Proc Natl Acad Sci USA. 2003;100:2890–5. doi: 10.1073/pnas.0437969100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Lehtonen E, Ordónez G, Reima I. Cytoskeleton in preimplantation mouse development. Cell Differ. 1988;24:165–77. doi: 10.1016/0045-6039(88)90048-6. [DOI] [PubMed] [Google Scholar]
  • 169.Moss DK, Bellett G, Carter JM, Liovic M, Keynton J, Prescott AR, Lane EB, Mogensen MM. Ninein is released from the centrosome and moves bi-directionally along microtubules. J Cell Sci. 2007;120:3064–74. doi: 10.1242/jcs.010322. [DOI] [PubMed] [Google Scholar]
  • 170.Duensing A, Liu Y, Perdreau SA, Kleylein-Sohn J, Nigg EA, Duensing S. Centriole overduplication through the concurrent formation of multiple daughter centrioles at single maternal templates. Oncogene. 2007;26:6280–8. doi: 10.1038/sj.onc.1210456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Spradling AC, Zheng Y. Developmental biology. The mother of all stem cells. Science. 2007;315:469–70. doi: 10.1126/science.1138237. [DOI] [PubMed] [Google Scholar]
  • 172.Singla V, Reiter JF. The primary cilium as the cell's antenna: signaling at a sensory organelle. Science. 2006;313:629–33. doi: 10.1126/science.1124534. [DOI] [PubMed] [Google Scholar]
  • 173.Kuang S, Kuroda K, Le Grand F, Rudnicki MA. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell. 2007;129:999–1010. doi: 10.1016/j.cell.2007.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Wilcock AC, Swedlow JR, Storey KG. Mitotic spindle orientation distinguishes stem cell and terminal modes of neuron production in the early spinal cord. Development. 2007;134:1943–54. doi: 10.1242/dev.002519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ciani L, Salinas PC. c-Jun N-terminal kinase (JNK) cooperates with Gsk3beta to regulate Dishevelled-mediated microtubule stability. BMC Cell Biol. 2007;8:27. doi: 10.1186/1471-2121-8-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kim Y, Zhou P, Qian L, Chuang JZ, Lee J, Li C, Iadecola C, Nathan C, Ding A. MyD88-5 links mitochondria, microtubules, and JNK3 in neurons and regulates neuronal survival. J Exp Med. 2007;204:2063–74. doi: 10.1084/jem.20070868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Smyth JT, Dehaven WI, Bird GS, Putney JW., Jr Role of the microtubule cytoskeleton in the function of the store-operated Ca2+ channel activator STIM1. J Cell Sci. 2007;120:3762–71. doi: 10.1242/jcs.015735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Petrache I, Birukova A, Ramirez SI, Garcia JG, Verin AD. The role of the microtubules in tumor necrosis factor-alpha-induced endothelial cell permeability. Am J Respir Cell Mol Biol. 2003;28:574–81. doi: 10.1165/rcmb.2002-0075OC. [DOI] [PubMed] [Google Scholar]
  • 179.Birukova AA, Birukov KG, Gorshkov B, Liu F, Garcia JG, Verin AD. MAP kinases in lung endothelial permeability induced by microtubule disassembly. Am J Physiol Lung Cell Mol Physiol. 2005;289:L75–84. doi: 10.1152/ajplung.00447.2004. [DOI] [PubMed] [Google Scholar]
  • 180.Hsieh CC, Papaconstantinou J. Thioredoxin-ASK1 complex levels regulate ROS-mediated p38 MAPK pathway activity in livers of aged and long-lived Snell dwarf mice. FASEB J. 2006;20:259–68. doi: 10.1096/fj.05-4376com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Lee CF, Liu CY, Hsieh RH, Wei YH. Oxidative stress-induced depolymerization of microtubules and alteration of mitochondrial mass in human cells. Ann N Y Acad Sci. 2005;1042:246–54. doi: 10.1196/annals.1338.027. [DOI] [PubMed] [Google Scholar]
  • 182.Banan A, Fields JZ, Decker H, Zhang Y, Keshavarzian A. Nitric oxide and its metabolites mediate ethanol-induced microtubule disruption and intestinal barrier dysfunction. J Pharmacol Exp Ther. 2000;294:997–1008. [PubMed] [Google Scholar]
  • 183.Kee SH, Steinert PM. Microtubule disruption in keratinocytes induces cell-cell adhesion through activation of endogenous E-cadherin. Mol Biol Cell. 2001;12:1983–93. doi: 10.1091/mbc.12.7.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Leung CG, Xu Y, Mularski B, Liu H, Gurbuxani S, Crispino JD. Requirements for survivin in terminal differentiation of erythroid cells and maintenance of hematopoietic stem and progenitor cells. J Exp Med. 2007;204:1603–11. doi: 10.1084/jem.20062395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, Endresen K, Ilebekk A, Mangschau A, Fjeld JG, Smith HJ, Taraldsrud E, Grøgaard HK, Bjørnerheim R, Brekke M, Müller C, Hopp E, Ragnarsson A, Brinchmann JE, Forfang K. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. NEJM. 2006;355:1199–1209. doi: 10.1056/NEJMoa055706. [DOI] [PubMed] [Google Scholar]
  • 186.Schächinger V, Erbs S, Elsässer A, Haberbosch W, Hambrecht R, Hölschermann H, Yu J, Corti R, Mathey DG, Hamm CW, Süselbeck T, Werner N, Haase J, Neuzner J, Germing A, Mark B, Assmus B, Tonn T, Dimmeler S, Zeiher AM, REPAIR-AMI Investigators. Improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: final 1-year results of the REPAIR-AMI trial. Eur Heart J. 2006;27:2775–83. doi: 10.1093/eurheartj/ehl388. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES