Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Stem Cell Res Ther ; 14(1): 322, 2023 11 08.
Article En | MEDLINE | ID: mdl-37941041

BACKGROUND: Cardiac pathological outcome of metabolic remodeling is difficult to model using cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) due to low metabolic maturation. METHODS: hiPSC-CM spheres were treated with AMP-activated protein kinase (AMPK) activators and examined for hiPSC-CM maturation features, molecular changes and the response to pathological stimuli. RESULTS: Treatment of hiPSC-CMs with AMPK activators increased ATP content, mitochondrial membrane potential and content, mitochondrial DNA, mitochondrial function and fatty acid uptake, indicating increased metabolic maturation. Conversely, the knockdown of AMPK inhibited mitochondrial maturation of hiPSC-CMs. In addition, AMPK activator-treated hiPSC-CMs had improved structural development and functional features-including enhanced Ca2+ transient kinetics and increased contraction. Transcriptomic, proteomic and metabolomic profiling identified differential levels of expression of genes, proteins and metabolites associated with a molecular signature of mature cardiomyocytes in AMPK activator-treated hiPSC-CMs. In response to pathological stimuli, AMPK activator-treated hiPSC-CMs had increased glycolysis, and other pathological outcomes compared to untreated cells. CONCLUSION: AMPK activator-treated cardiac spheres could serve as a valuable model to gain novel insights into cardiac diseases.


AMP-Activated Protein Kinases , Induced Pluripotent Stem Cells , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cells, Cultured , Proteomics , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/physiology
2.
Nat Metab ; 1(3): 390-403, 2019 03.
Article En | MEDLINE | ID: mdl-31535081

Amino acid (AA) metabolism is involved in diverse cellular functions, including cell survival and growth, however it remains unclear how it regulates normal hematopoiesis versus leukemogenesis. Here, we report that knockout of Slc1a5 (ASCT2), a transporter of neutral AAs, especially glutamine, results in mild to moderate defects in bone marrow and mature blood cell development under steady state conditions. In contrast, constitutive or induced deletion of Slc1a5 decreases leukemia initiation and maintenance driven by the oncogene MLL-AF9 or Pten deficiency. Survival of leukemic mice is prolonged following Slc1a5 deletion, and pharmacological inhibition of ASCT2 also decreases leukemia development and progression in xenograft models of human acute myeloid leukemia. Mechanistically, loss of ASCT2 generates a global effect on cellular metabolism, disrupts leucine influx and mTOR signaling, and induces apoptosis in leukemic cells. Given the substantial difference in reliance on ASCT2-mediated AA metabolism between normal and malignant blood cells, this in vivo study suggests ASCT2 as a promising therapeutic target for the treatment of leukemia.


Amino Acid Transport System ASC/physiology , Amino Acids/metabolism , Leukemia, Myeloid, Acute/metabolism , Minor Histocompatibility Antigens/physiology , Amino Acid Transport System ASC/genetics , Animals , Cell Line, Tumor , Disease Progression , Hematopoiesis/genetics , Heterografts , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Minor Histocompatibility Antigens/genetics
3.
J Mol Cell Cardiol ; 132: 120-135, 2019 07.
Article En | MEDLINE | ID: mdl-31082397

Immature phenotypes of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) limit the utility of these cells in clinical application and basic research. During cardiac development, postnatal cardiomyocytes experience high oxygen tension along with a concomitant downregulation of hypoxia-inducible factor 1α (HIF-1α), leading to increased fatty acid oxidation (FAO). We hypothesized that targeting HIF-1α alone or in combination with other metabolic regulators could promote the metabolic maturation of hiPSC-CMs. We examined the effect of HIF-1α inhibition on the maturation of hiPSC-CMs and investigated a multipronged approach to promote hiPSC-CM maturation by combining HIF-1α inhibition with molecules that target key pathways involved in the energy metabolism. Cardiac spheres of highly-enriched hiPSC-CMs were treated with a HIF-1α inhibitor alone or in combination with an agonist of peroxisome proliferator activated receptor α (PPARα) and three postnatal factors (triiodothyronine hormone T3, insulin-like growth factor-1 and dexamethasone). HIF-1α inhibition significantly increased FAO and basal and maximal respiration of hiPSC-CMs. Combining HIF-1α inhibition with PPARα activation and the postnatal factors further increased FAO and improved mitochondrial maturation in hiPSC-CMs. Compared with mock-treated cultures, the cultures treated with the five factors had increased mitochondrial content and contained more cells with mitochondrial distribution throughout the cells, which are features of more mature cardiomyocytes. Consistent with these observations, a number of transcriptional regulators of mitochondrial metabolic processes were upregulated in hiPSC-CMs treated with the five factors. Furthermore, these cells had significantly increased Ca2+ transient kinetics and contraction and relaxation velocities, which are functional features for more mature cardiomyocytes. Therefore, targeting HIF-1α in combination with other metabolic regulators significantly improves the metabolic maturation of hiPSC-CMs.


Benzamides/pharmacology , Drug Synergism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Induced Pluripotent Stem Cells/physiology , Mitochondria/metabolism , Myocytes, Cardiac/physiology , PPAR alpha/agonists , Anti-Inflammatory Agents/pharmacology , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Dexamethasone/pharmacology , Energy Metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Insulin-Like Growth Factor I/pharmacology , Lipid Metabolism , Mitochondria/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Transcriptome , Triiodothyronine/pharmacology
4.
Cell Stem Cell ; 24(4): 608-620.e6, 2019 04 04.
Article En | MEDLINE | ID: mdl-30880025

Hematopoietic stem cell (HSC) quiescence is a tightly regulated process crucial for hematopoietic regeneration, which requires a healthy and supportive microenvironmental niche within the bone marrow (BM). Here, we show that deletion of Ptpn21, a protein tyrosine phosphatase highly expressed in HSCs, induces stem cell egress from the niche due to impaired retention within the BM. Ptpn21-/- HSCs exhibit enhanced mobility, decreased quiescence, increased apoptosis, and defective reconstitution capacity. Ptpn21 deletion also decreased HSC stiffness and increased physical deformability, in part by dephosphorylating Spetin1 (Tyr246), a poorly described component of the cytoskeleton. Elevated phosphorylation of Spetin1 in Ptpn21-/- cells impaired cytoskeletal remodeling, contributed to cortical instability, and decreased cell rigidity. Collectively, these findings show that Ptpn21 maintains cellular mechanics, which is correlated with its important functions in HSC niche retention and preservation of hematopoietic regeneration capacity.


Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Septins/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Stem Cell Niche
5.
Sci Adv ; 4(10): eaat2681, 2018 10.
Article En | MEDLINE | ID: mdl-30338292

While deregulation of mitochondrial metabolism and cytosolic glycolysis has been well recognized in tumor cells, the role of coordinated mitochondrial oxidation and cytosolic fermentation of pyruvate, a key metabolite derived from glucose, in physiological processes is not well understood. Here, we report that knockout of PTPMT1, a mitochondrial phosphoinositide phosphatase, completely blocked postnatal cerebellar development. Proliferation of granule cell progenitors, the most actively replicating cells in the developing cerebellum, was only moderately decreased, and proliferation of Purkinje cell progenitors did not seem to be affected in knockout mice. In contrast, generation of functional Bergmann glia from multipotent precursor cells (radial glia), which is essential for cerebellar corticogenesis, was totally disrupted. Moreover, despite a low turnover rate, neural stem cells were impaired in self-renewal in knockout mice. Mechanistically, loss of PTPMT1 decreased mitochondrial aerobic metabolism by limiting utilization of pyruvate, which resulted in bioenergetic stress in neural precursor/stem cells but not in progenitor or mature cells, leading to cell cycle arrest through activation of the AMPK-p19/p21 pathway. This study suggests that mitochondrial oxidation of the carbohydrate fuel is required for postnatal cerebellar development, and identifies a bioenergetic stress-induced cell cycle checkpoint in neural precursor/stem cells.


Carbohydrate Metabolism/physiology , Cerebellum/growth & development , Cerebellum/metabolism , Mitochondria/metabolism , Neural Stem Cells/physiology , Animals , Animals, Newborn , Cell Cycle Checkpoints/genetics , Cerebellum/cytology , Female , Glycolysis , Male , Mice, Knockout , Neural Stem Cells/cytology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pyruvic Acid/metabolism
6.
Oncotarget ; 9(31): 21831-21843, 2018 Apr 24.
Article En | MEDLINE | ID: mdl-29774106

Juvenile Myelomonocytic Leukemia (JMML) is a pediatric myeloproliferative neoplasm (MPN) that has a poor prognosis. Somatic mutations in Ptpn11 are the most frequent cause of JMML and they commonly occur in utero. Animal models of mutant Ptpn11 have probed the signaling pathways that contribute to JMML. However, existing models may inappropriately exacerbate MPN features by relying on non-hematopoietic-restricted Cre-loxP strains or transplantations into irradiated recipients. In this study we generate hematopoietic-restricted models of Ptpn11E76K-mediated disease using Csf1r-MCM and Flt3Cre. We show that these animals have indolent MPN progression despite robust GM-CSF hypersensitivity and Ras-Erk hyperactivation. Rather, the dominant pathology is pronounced thrombocytopenia with expanded extramedullary hematopoiesis. Furthermore, we demonstrate that the timing of tamoxifen administration in Csf1r-MCM mice can specifically induce recombinase activity in either fetal or adult hematopoietic progenitors. We take advantage of this technique to show more rapid monocytosis following Ptpn11E76K expression in fetal progenitors compared with adult progenitors. Finally, we demonstrate that Ptpn11E76K results in the progressive reduction of T cells, most notably of CD4+ and naïve T cells. This corresponds to an increased frequency of T cell progenitors in the thymus and may help explain the occasional emergence of T-cell leukemias in JMML patients. Overall, our study is the first to describe the consequences of hematopoietic-restricted Ptpn11E76K expression in the absence of irradiation. Our techniques can be readily adapted by other researchers studying somatically-acquired blood disorders.

7.
Sci Signal ; 11(522)2018 03 20.
Article En | MEDLINE | ID: mdl-29559584

Catalytically activating mutations in Ptpn11, which encodes the protein tyrosine phosphatase SHP2, cause 50% of Noonan syndrome (NS) cases, whereas inactivating mutations in Ptpn11 are responsible for nearly all cases of the similar, but distinct, developmental disorder Noonan syndrome with multiple lentigines (NSML; formerly called LEOPARD syndrome). However, both types of disease mutations are gain-of-function mutations because they cause SHP2 to constitutively adopt an open conformation. We found that the catalytic activity of SHP2 was required for the pathogenic effects of gain-of-function, disease-associated mutations on the development of hydrocephalus in the mouse. Targeted pan-neuronal knockin of a Ptpn11 allele encoding the active SHP2 E76K mutant resulted in hydrocephalus due to aberrant development of ependymal cells and their cilia. These pathogenic effects of the E76K mutation were suppressed by the additional mutation C459S, which abolished the catalytic activity of SHP2. Moreover, ependymal cells in NSML mice bearing the inactive SHP2 mutant Y279C were also unaffected. Mechanistically, the SHP2 E76K mutant induced developmental defects in ependymal cells by enhancing dephosphorylation and inhibition of the transcription activator STAT3. Whereas STAT3 activity was reduced in Ptpn11E76K/+ cells, the activities of the kinases ERK and AKT were enhanced, and neural cell-specific Stat3 knockout mice also manifested developmental defects in ependymal cells and cilia. These genetic and biochemical data demonstrate a catalytic-dependent role of SHP2 gain-of-function disease mutants in the pathogenesis of hydrocephalus.


Gain of Function Mutation , Hydrocephalus/genetics , LEOPARD Syndrome/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Animals , Biocatalysis , Ependyma/cytology , Ependyma/metabolism , Genetic Predisposition to Disease/genetics , Humans , Hydrocephalus/metabolism , LEOPARD Syndrome/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/metabolism , Noonan Syndrome/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
8.
J Clin Invest ; 128(3): 944-959, 2018 03 01.
Article En | MEDLINE | ID: mdl-29376892

Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMß2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.


Factor XII/metabolism , Neutrophils/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Wound Healing , Animals , Calcium/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Extracellular Traps , Female , Humans , Inflammation , Leukocytes/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
9.
Mol Cell ; 69(1): 87-99.e7, 2018 01 04.
Article En | MEDLINE | ID: mdl-29249655

Loss of LKB1 is associated with increased metastasis and poor prognosis in lung cancer, but the development of targeted agents is in its infancy. Here we report that a glutaminolytic enzyme, glutamate dehydrogenase 1 (GDH1), upregulated upon detachment via pleomorphic adenoma gene 1 (PLAG1), provides anti-anoikis and pro-metastatic signals in LKB1-deficient lung cancer. Mechanistically, the GDH1 product α-KG activates CamKK2 by enhancing its substrate AMPK binding, which contributes to energy production that confers anoikis resistance. The effect of GDH1 on AMPK is evident in LKB1-deficient lung cancer, where AMPK activation predominantly depends on CamKK2. Targeting GDH1 with R162 attenuated tumor metastasis in patient-derived xenograft model and correlation studies in lung cancer patients further validated the clinical relevance of our finding. Our study provides insight into the molecular mechanism by which GDH1-mediated metabolic reprogramming of glutaminolysis mediates lung cancer metastasis and offers a therapeutic strategy for patients with LKB1-deficient lung cancer.


Anoikis/physiology , DNA-Binding Proteins/metabolism , Glutamate Dehydrogenase/metabolism , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Small Cell Lung Carcinoma/pathology , A549 Cells , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line, Tumor , Enzyme Activation/physiology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Transplantation, Heterologous
10.
Nature ; 539(7628): 304-308, 2016 11 10.
Article En | MEDLINE | ID: mdl-27783593

Germline activating mutations of the protein tyrosine phosphatase SHP2 (encoded by PTPN11), a positive regulator of the RAS signalling pathway, are found in 50% of patients with Noonan syndrome. These patients have an increased risk of developing leukaemia, especially juvenile myelomonocytic leukaemia (JMML), a childhood myeloproliferative neoplasm (MPN). Previous studies have demonstrated that mutations in Ptpn11 induce a JMML-like MPN through cell-autonomous mechanisms that are dependent on Shp2 catalytic activity. However, the effect of these mutations in the bone marrow microenvironment remains unclear. Here we report that Ptpn11 activating mutations in the mouse bone marrow microenvironment promote the development and progression of MPN through profound detrimental effects on haematopoietic stem cells (HSCs). Ptpn11 mutations in mesenchymal stem/progenitor cells and osteoprogenitors, but not in differentiated osteoblasts or endothelial cells, cause excessive production of the CC chemokine CCL3 (also known as MIP-1α), which recruits monocytes to the area in which HSCs also reside. Consequently, HSCs are hyperactivated by interleukin-1ß and possibly other proinflammatory cytokines produced by monocytes, leading to exacerbated MPN and to donor-cell-derived MPN following stem cell transplantation. Remarkably, administration of CCL3 receptor antagonists effectively reverses MPN development induced by the Ptpn11-mutated bone marrow microenvironment. This study reveals the critical contribution of Ptpn11 mutations in the bone marrow microenvironment to leukaemogenesis and identifies CCL3 as a potential therapeutic target for controlling leukaemic progression in Noonan syndrome and for improving stem cell transplantation therapy in Noonan-syndrome-associated leukaemias.


Cell Transformation, Neoplastic/genetics , Cellular Microenvironment/genetics , Hematopoietic Stem Cells/pathology , Leukemia/genetics , Leukemia/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Stem Cell Niche/genetics , Animals , Chemokine CCL3/antagonists & inhibitors , Chemokine CCL3/metabolism , Disease Progression , Endothelial Cells/cytology , Female , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-1beta/metabolism , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Monocytes/metabolism , Mutation , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Noonan Syndrome/pathology , Osteoblasts/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Stem Cell Transplantation
11.
Leuk Res ; 50: 132-140, 2016 11.
Article En | MEDLINE | ID: mdl-27760406

Current therapy for acute myeloid leukemia (AML) primarily includes high-dose cytotoxic chemotherapy with or without allogeneic stem cell transplantation. Targeting unique cellular metabolism of cancer cells is a potentially less toxic approach. Monotherapy with mitochondrial inhibitors like metformin have met with limited success since escape mechanisms such as increased glycolytic ATP production, especially in hyperglycemia, can overcome the metabolic blockade. As an alternative strategy for metformin therapy, we hypothesized that the combination of 6-benzylthioinosine (6-BT), a broad-spectrum metabolic inhibitor, and metformin could block this drug resistance mechanism. Metformin treatment alone resulted in significant suppression of ROS and mitochondrial respiration with increased glycolysis accompanied by modest cytotoxicity (10-25%). In contrast, 6-BT monotherapy resulted in inhibition of glucose uptake, decreased glycolysis, and decreased ATP with minimal changes in ROS and mitochondrial respiration. The combination of 6-BT with metformin resulted in significant cytotoxicity (60-70%) in monocytic AML cell lines and was associated with inhibition of FLT3-ITD activated STAT5 and reduced c-Myc and GLUT-1 expression. Therefore, although the anti-tumor and metabolic effects of metformin have been limited by the metabolic reprogramming within cells, the novel combination of 6-BT and metformin targets this bypass mechanism resulting in reduced glycolysis, STAT5 inhibition, and increased cell death.


Cell Death/drug effects , Leukemia, Myeloid, Acute/drug therapy , Metformin/therapeutic use , Thioinosine/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Synergism , Fetal Blood/cytology , Glycolysis/drug effects , Humans , Inverted Repeat Sequences , Leukemia, Myeloid, Acute/genetics , STAT5 Transcription Factor/antagonists & inhibitors , Thioinosine/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/physiology
12.
Blood ; 125(10): 1562-5, 2015 Mar 05.
Article En | MEDLINE | ID: mdl-25593337

The difficulty in maintaining the reconstituting capabilities of hematopoietic stem cells (HSCs) in culture outside of the bone marrow microenvironment has severely limited their utilization for clinical therapy. This hurdle is largely due to the differentiation of long-term stem cells. Emerging evidence suggests that energy metabolism plays an important role in coordinating HSC self-renewal and differentiation. Here, we show that treatment with alexidine dihydrochloride, an antibiotic and a selective inhibitor of the mitochondrial phosphatase Ptpmt1, which is crucial for the differentiation of HSCs, reprogrammed cellular metabolism from mitochondrial aerobic metabolism to glycolysis, resulting in a remarkable preservation of long-term HSCs ex vivo in part through hyperactivation of adenosine 5'-monophosphate-activated protein kinase (AMPK). In addition, inhibition of mitochondrial metabolism and activation of AMPK by metformin, a diabetes drug, also decreased differentiation and helped maintain stem cells in culture. Thus, manipulating metabolic pathways represents an effective new strategy for ex vivo maintenance of HSCs.


Biguanides/pharmacology , Cellular Reprogramming/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Aerobiosis/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glycolysis/drug effects , Hematopoietic Stem Cells/cytology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mice , Oxygen Consumption/drug effects
13.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Article En | MEDLINE | ID: mdl-24265312

CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.


Embryonic Stem Cells/metabolism , Glycolysis/physiology , Mitochondria/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic/physiology , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Hypoxia/physiology , Embryonic Stem Cells/cytology , Glucose/genetics , Glucose/metabolism , Hexokinase/biosynthesis , Hexokinase/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidation-Reduction , Repressor Proteins/genetics , Trans-Activators/genetics
14.
Mol Cancer Ther ; 12(9): 1738-48, 2013 Sep.
Article En | MEDLINE | ID: mdl-23825065

Activating mutations in PTPN11 (encoding SHP2), a protein tyrosine phosphatase (PTP) that plays an overall positive role in growth factor and cytokine signaling, are directly associated with the pathogenesis of Noonan syndrome and childhood leukemias. Identification of SHP2-selective inhibitors could lead to the development of new drugs that ultimately serve as treatments for PTPN11-associated diseases. As the catalytic core of SHP2 shares extremely high homology to those of SHP1 and other PTPs that play negative roles in cell signaling, to identify selective inhibitors of SHP2 using computer-aided drug design, we targeted a protein surface pocket that is adjacent to the catalytic site, is predicted to be important for binding to phosphopeptide substrates, and has structural features unique to SHP2. From computationally selected candidate compounds, #220-324 effectively inhibited SHP2 activity with an IC50 of 14 µmol/L. Fluorescence titration experiments confirmed its direct binding to SHP2. This active compound was further verified for its ability to inhibit SHP2-mediated cell signaling and cellular function with minimal off-target effects. Furthermore, mouse myeloid progenitors with the activating mutation (E76K) in PTPN11 and patient leukemic cells with the same mutation were more sensitive to this inhibitor than wild-type cells. This study provides evidence that SHP2 is a "druggable" target for the treatment of PTPN11-associated diseases. As the small-molecule SHP2 inhibitor identified has a simple chemical structure, it represents an ideal lead compound for the development of novel anti-SHP2 drugs. Mol Cancer Ther; 12(9); 1738-48. ©2013 AACR.


Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Leukemia, Myeloid/pathology , Noonan Syndrome/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Triazines/pharmacology , Animals , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Line , Child , Drug Design , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Enzyme Inhibitors/analysis , Enzyme Inhibitors/chemistry , Gene Knockout Techniques , Humans , Indoles/chemistry , Indoles/metabolism , Leukemia, Myeloid/drug therapy , Mice , Molecular Structure , Mutation , Noonan Syndrome/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triazines/chemistry , Triazines/metabolism , Tumor Cells, Cultured
15.
PLoS One ; 8(5): e63152, 2013.
Article En | MEDLINE | ID: mdl-23675459

Gain of function (GOF) mutations in protein tyrosine phosphatase Ptpn11 have been identified in childhood leukemias, and these mutations are sufficient to drive the development of myeloproliferative disorder and malignant leukemias in mice. However, the molecular mechanisms by which Ptpn11 mutations induce these malignancies are not completely understood. Here we report that Ptpn11 GOF mutations cause cytokine hypersensitivity in hematopoietic cells partly by enhancing the production of reactive oxygen species (ROS). GOF mutations D61G or E76K in Ptpn11 increased ROS levels in myeloid progenitors but not in hematopoietic stem cells. Increased ROS enhanced cellular responses to cytokines by promoting cytokine signaling. Treatment with an antioxidant partially corrected cytokine hypersensitivity in Ptpn11 mutant progenitors. Further analyses demonstrated that Ptpn11 mutations increased mitochondrial aerobic metabolism by interacting with a novel substrate in the mitochondria. This study provides new insights into the pathogenic effects of GOF mutations of Ptpn11 and implies that antioxidants may have a therapeutic benefit for the leukemic patients with these mutations.


Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cytokines/metabolism , Disease Models, Animal , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Myeloid Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction
16.
Cell Stem Cell ; 12(1): 62-74, 2013 Jan 03.
Article En | MEDLINE | ID: mdl-23290137

The regulation and coordination of mitochondrial metabolism with hematopoietic stem cell (HSC) self-renewal and differentiation is not fully understood. Here we report that depletion of PTPMT1, a PTEN-like mitochondrial phosphatase, in inducible or hematopoietic-cell-specific knockout mice resulted in hematopoietic failure due to changes in the cell cycle and a block in the differentiation of HSCs. Surprisingly, the HSC pool was increased by âˆ¼40-fold in PTPMT1 knockout mice. Reintroduction of wild-type PTPMT1, but not catalytically deficient PTPMT1 or truncated PTPMT1 lacking mitochondrial localization, restored differentiation capabilities of PTPMT1 knockout HSCs. Further analyses demonstrated that PTPMT1 deficiency altered mitochondrial metabolism and that phosphatidylinositol phosphate substrates of PTPMT1 directly enhanced fatty-acid-induced activation of mitochondrial uncoupling protein 2. Intriguingly, depletion of PTPMT1 from myeloid, T lymphoid, or B lymphoid progenitors did not cause any defects in lineage-specific knockout mice. This study establishes a crucial role of PTPMT1 in the metabolic regulation of HSC function.


Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cells, Cultured , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , PTEN Phosphohydrolase/genetics , Uncoupling Protein 2
17.
Article Zh | MEDLINE | ID: mdl-22164631

A schistosomiasis surveillance was performed with the Oncomelania snail survey and suspected patient survey in Jiaxing City, 2009. There were snails in 3 villages of 2 counties and the airport of Jiaxing with the area of 17 390 m2, and the highest density was 64 snails per 0.1 m2. A total of 2 368 snails were dissected, and no infected snail was found. One case of acute schistosomiasis was found and the patient was an immigrant worker from Yueyang City, Hunan Province. In conclusion, there is still a threat of schistosomiasis in Jiaxing City.


Disease Reservoirs/parasitology , Schistosomiasis/epidemiology , Schistosomiasis/prevention & control , Sentinel Surveillance , Snails/growth & development , Adolescent , Adult , Aged , Animals , Child , China/epidemiology , Feces/parasitology , Female , Humans , Male , Middle Aged , Pest Control , Schistosoma/isolation & purification , Schistosoma/physiology , Schistosomiasis/parasitology , Snails/parasitology , Young Adult
18.
Mol Cell Biol ; 31(24): 4902-16, 2011 Dec.
Article En | MEDLINE | ID: mdl-21986498

Mitochondria are highly dynamic organelles that play multiple roles in cells. How mitochondria cooperatively modulate embryonic stem (ES) cell function during development is not fully understood. Global disruption of Ptpmt1, a mitochondrial Pten-like phosphatidylinositol phosphate (PIP) phosphatase, resulted in developmental arrest and postimplantation lethality. Ptpmt1(-/-) blastocysts failed to outgrow, and inner-cell-mass cells failed to thrive. Depletion of Ptpmt1 in conditional knockout ES cells decreased proliferation without affecting energy homeostasis or cell survival. Differentiation of Ptpmt1-depleted ES cells was essentially blocked. This was accompanied by upregulation of cyclin-dependent kinase inhibitors and a significant cell cycle delay. Reintroduction of wild-type but not of catalytically deficient Ptpmt1 C132S or truncated Ptpmt1 lacking the mitochondrial localization signal restored the differentiation capabilities of Ptpmt1 knockout ES cells. Intriguingly, Ptpmt1 is specifically important for stem cells, as ablation of Ptpmt1 in differentiated embryonic fibroblasts did not disturb cellular function. Further analyses demonstrated that oxygen consumption of Ptpmt1-depleted cells was decreased, while glycolysis was concomitantly enhanced. In addition, mitochondrial fusion/dynamics were compromised in Ptpmt1 knockout cells due to accumulation of PIPs. These studies, while establishing a crucial role for Ptpmt1 phosphatase in embryogenesis, reveal a mitochondrial metabolic stress-activated checkpoint in the control of ES cell differentiation.


Cell Cycle Checkpoints , Cell Differentiation , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , Alleles , Animals , Apoptosis , Cell Proliferation , Cloning, Molecular , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Glycolysis , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Knockout , Microscopy, Confocal , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/metabolism , Reactive Oxygen Species/analysis , Stress, Physiological
19.
J Exp Med ; 208(10): 1977-88, 2011 Sep 26.
Article En | MEDLINE | ID: mdl-21930766

Activating mutations in protein tyrosine phosphatase 11 (Ptpn11) have been identified in childhood acute leukemias, in addition to juvenile myelomonocytic leukemia (JMML), which is a myeloproliferative disorder (MPD). It is not clear whether activating mutations of this phosphatase play a causal role in the pathogenesis of acute leukemias. If so, the cell origin of leukemia-initiating stem cells (LSCs) remains to be determined. Ptpn11(E76K) mutation is the most common and most active Ptpn11 mutation found in JMML and acute leukemias. However, the pathogenic effects of this mutation have not been well characterized. We have created Ptpn11(E76K) conditional knock-in mice. Global Ptpn11(E76K/+) mutation results in early embryonic lethality. Induced knock-in of this mutation in pan hematopoietic cells leads to MPD as a result of aberrant activation of hematopoietic stem cells (HSCs) and myeloid progenitors. These animals subsequently progress to acute leukemias. Intriguingly, in addition to acute myeloid leukemia (AML), T cell acute lymphoblastic leukemia/lymphoma (T-ALL) and B-ALL are evolved. Moreover, tissue-specific knock-in of Ptpn11(E76K/+) mutation in lineage-committed myeloid, T lymphoid, and B lymphoid progenitors also results in AML, T-ALL, and B-ALL, respectively. Further analyses have revealed that Shp2 (encoded by Ptpn11) is distributed to centrosomes and that Ptpn11(E76K/+) mutation promotes LSC development, partly by causing centrosome amplification and genomic instability. Thus, Ptpn11(E76K) mutation has non-lineage-specific effects on malignant transformation of hematopoietic cells and initiates acute leukemias at various stages of hematopoiesis.


Cell Transformation, Neoplastic , Hematopoietic Stem Cells/physiology , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Animals , Cell Differentiation/physiology , Cell Lineage , Centrosome/metabolism , Chromosomal Instability , Embryo, Mammalian/pathology , Embryo, Mammalian/physiology , Gene Knock-In Techniques , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
20.
Blood ; 116(18): 3611-21, 2010 Nov 04.
Article En | MEDLINE | ID: mdl-20651068

Germline and somatic gain-of-function mutations in tyrosine phosphatase PTPN11 (SHP-2) are associated with juvenile myelomonocytic leukemia (JMML), a myeloproliferative disease (MPD) of early childhood. The mechanism by which PTPN11 mutations induce this disease is not fully understood. Signaling partners that mediate the pathogenic effects of PTPN11 mutations have not been explored. Here we report that germ line mutation Ptpn11(D61G) in mice aberrantly accelerates hematopoietic stem cell (HSC) cycling, increases the stem cell pool, and elevates short-term and long-term repopulating capabilities, leading to the development of MPD. MPD is reproduced in primary and secondary recipient mice transplanted with Ptpn11(D61G/+) whole bone marrow cells or purified Lineage(-)Sca-1(+)c-Kit(+) cells, but not lineage committed progenitors. The deleterious effects of Ptpn11(D61G) mutation on HSCs are attributable to enhancing cytokine/growth factor signaling. The aberrant HSC activities caused by Ptpn11(D61G) mutation are largely corrected by deletion of Gab2, a prominent interacting protein and target of Shp-2 in cell signaling. As a result, MPD phenotypes are markedly ameliorated in Ptpn11(D61G/+)/Gab2(-/-) double mutant mice. Collectively, our data suggest that oncogenic Ptpn11 induces MPD by aberrant activation of HSCs. This study also identifies Gab2 as an important mediator for the pathogenic effects of Ptpn11 mutations.


Hematopoietic Stem Cells/pathology , Mutation , Myeloproliferative Disorders/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Interleukin-3/immunology , Mice , Mice, Inbred C57BL , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phosphoproteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
...