Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
Mol Cell ; 69(1): 87-99.e7, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29249655

ABSTRACT

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.


Subject(s)
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
2.
Nature ; 539(7628): 304-308, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27783593

ABSTRACT

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.


Subject(s)
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
3.
Curr Opin Hematol ; 27(4): 288-293, 2020 07.
Article in English | MEDLINE | ID: mdl-32487806

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to summarize the current understanding of germline mutations as they contribute to leukemia development and progression. We also discuss how these new insights may help improve clinical management of germline mutations associated with leukemia. RECENT FINDINGS: Germline mutations may represent important initial mutations in the development of leukemia where interaction with somatic mutations provide further hits in leukemic progression. In addition, germline mutations may also contribute to leukemogenesis by impacting bone marrow stem-cell microenvironment and immune cell development and function. SUMMARY: Leukemia is characterized by the clonal expansion of malignant cells secondary to somatic or germline mutations in a variety of genes. Understanding somatic mutations that drive leukemogenesis has drastically improved our knowledge of leukemia biology and led to novel therapeutic strategies. Advances have also been made in identifying germline mutations that may affect leukemic development and progression. This review will discuss the biological and clinical relationship of germline mutations with clonal hematopoiesis, bone marrow microenvironment, and immunity in the progression of leukemia.


Subject(s)
Bone Marrow Cells , Carcinogenesis , Germ-Line Mutation , Leukemia , Neoplastic Stem Cells , Stem Cell Niche/genetics , Tumor Microenvironment/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Humans , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
4.
J Pathol ; 247(1): 135-146, 2019 01.
Article in English | MEDLINE | ID: mdl-30376595

ABSTRACT

The Src homology-2 domain-containing tyrosine phosphatase 2 (SHP-2) regulates many cellular processes, including proliferation, differentiation and survival. Polymorphisms in the gene encoding SHP-2 are associated with an increased susceptibility to develop ulcerative colitis. We recently reported that intestinal epithelial cell (IEC)-specific deletion of Shp-2 in mice (Shp-2IEC-KO ) leads to chronic colitis and colitis-associated cancer. This suggests that SHP-2-dependent signaling protects the colonic epithelium against inflammation and colitis-associated cancer development. To verify this hypothesis, we generated mice expressing the Shp-2 E76K activated form specifically in IEC. Our results showed that sustained Shp-2 activation in IEC increased intestine and crypt length, correlating with increased cell proliferation and migration. Crypt regeneration capacity was also markedly enhanced, as revealed by ex vivo organoid culture. Shp-2 activation alters the secretory cell lineage, as evidenced by increased goblet cell numbers and mucus secretion. Notably, these mice also demonstrated elevated ERK signaling in IEC and exhibited resistance against both chemical- and Citrobacter rodentium-induced colitis. In contrast, mice with IEC-specific Shp-2 deletion displayed reduced ERK signaling and rapidly developed chronic colitis. Remarkably, expression of an activated form of Braf in Shp-2-deficient mice restored ERK activation, goblet cell production and prevented colitis. Altogether, our results indicate that chronic activation of Shp-2/ERK signaling in the colonic epithelium confers resistance to mucosal erosion and colitis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Movement , Cell Proliferation , Colitis/prevention & control , Colon/enzymology , Goblet Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Regeneration , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Colitis/enzymology , Colitis/genetics , Colitis/pathology , Colon/pathology , Disease Models, Animal , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Goblet Cells/pathology , Mice, Transgenic , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Tissue Culture Techniques , Wound Healing
5.
J Mol Cell Cardiol ; 132: 120-135, 2019 07.
Article in English | MEDLINE | ID: mdl-31082397

ABSTRACT

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.


Subject(s)
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
6.
Proc Natl Acad Sci U S A ; 113(4): 984-9, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755576

ABSTRACT

Gain-of-function (GOF) mutations of protein tyrosine phosphatase nonreceptor type 11 Ptpn11 (Shp2), a protein tyrosine phosphatase implicated in multiple cell signaling pathways, are associated with childhood leukemias and solid tumors. The underlying mechanisms are not fully understood. Here, we report that Ptpn11 GOF mutations disturb mitosis and cytokinesis, causing chromosomal instability and greatly increased susceptibility to DNA damage-induced malignancies. We find that Shp2 is distributed to the kinetochore, centrosome, spindle midzone, and midbody, all of which are known to play critical roles in chromosome segregation and cytokinesis. Mouse embryonic fibroblasts with Ptpn11 GOF mutations show a compromised mitotic checkpoint. Centrosome amplification and aberrant mitosis with misaligned or lagging chromosomes are significantly increased in Ptpn11-mutated mouse and patient cells. Abnormal cytokinesis is also markedly increased in these cells. Further mechanistic analyses reveal that GOF mutant Shp2 hyperactivates the Polo-like kinase 1 (Plk1) kinase by enhancing c-Src kinase-mediated tyrosine phosphorylation of Plk1. This study provides novel insights into the tumorigenesis associated with Ptpn11 GOF mutations and cautions that DNA-damaging treatments in Noonan syndrome patients with germ-line Ptpn11 GOF mutations could increase the risk of therapy-induced malignancies.


Subject(s)
DNA Damage , Mitosis , Neoplasms/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Cell Cycle Proteins/metabolism , Chromosomal Instability , Humans , Mice , Mice, Inbred C57BL , Mutation , Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
7.
Blood ; 125(10): 1562-5, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25593337

ABSTRACT

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.


Subject(s)
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
8.
Curr Opin Hematol ; 23(4): 339-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27071022

ABSTRACT

PURPOSE OF REVIEW: Hematopoietic stem cells (HSCs) are a population of cells in the bone marrow which can self-renew, differentiate into late lineage progenitors, or remain quiescent. HSCs exist alongside several cell types in the bone marrow microenvironment that comprise the stem cell niche. These cells regulate HSC function and can contribute to leukemogenesis. In this review we will discuss recent advances in this field. RECENT FINDINGS: In the vascular niche, arteriolar and sinusoidal zones appear to play distinct roles in HSC function. Endothelial cells modulate HSC function via Notch and other signaling pathways. In the endosteal niche multiple cell types regulate HSCs. Osteoblasts promote HSC quiescence via secreted factors and possibly physical interactions, whereas adipocytes may oppose HSC quiescence. The balance of these opposing factors depends on metabolic cues. Feedback from HSC-derived cells, including macrophages and megakaryocytes also appears to regulate HSC quiescence. Dysfunction of the bone marrow microenvironment, including mesenchymal stem cell-derived stromal cells and the sympathetic nervous system can induce or alter the progression of hematologic malignancies. SUMMARY: Many cell types in the bone marrow microenvironment affect HSC function and contribute to malignancy. Further understanding how HSCs are regulated by the microenvironment has clinical implications for stem cell transplantation and other therapies for hematologic malignancies.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cellular Microenvironment , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukemia/etiology , Leukemia/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Cycle , Cell Differentiation , Cell Self Renewal , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , Hematologic Neoplasms/etiology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Leukemia/pathology , Signal Transduction , Stem Cell Niche
9.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24265312

ABSTRACT

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.


Subject(s)
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
10.
J Biol Chem ; 288(36): 25727-25738, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23884424

ABSTRACT

Activating mutations in Ptpn11 (Shp2), a protein tyrosine phosphatase involved in diverse cell signaling pathways, are associated with pediatric leukemias and solid tumors. However, the pathogenic effects of these mutations have not been fully characterized. Here, we report that induction of the Ptpn11(E76K/+) mutation, the most common and active Ptpn11 mutation found in leukemias and solid tumors, in primary mouse embryonic fibroblasts resulted in proliferative arrest and premature senescence. As a result, apoptosis was markedly increased. These cellular responses were accompanied and mediated by up-regulation of p53 and p21. Moreover, intracellular levels of reactive oxygen species (ROS), byproducts of mitochondrial oxidative phosphorylation, were elevated in Ptpn11(E76K/+) cells. Since Shp2 is also distributed to the mitochondria (in addition to the cytosol), the impact of the Ptpn11(E76K/+) mutation on mitochondrial function was analyzed. These analyses revealed that oxygen consumption of Ptpn11(E76K/+) cells and the respiratory function of Ptpn11(E76K/+) mitochondria were significantly increased. Furthermore, we found that phosphorylation of mitochondrial Stat3, one of the substrates of Shp2 phosphatase, was greatly decreased in the mutant cells with the activating mutation Ptpn11(E76K/+). This study provides novel insights into the initial effects of tumor-associated Ptpn11 mutations.


Subject(s)
Cellular Senescence , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Mutation, Missense , Neoplasms/enzymology , Oxidative Stress , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Oxygen Consumption/genetics , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Mol Cell Biochem ; 392(1-2): 31-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24623267

ABSTRACT

MTMR14 is a phosphoinositide phosphatase, which has been reported to regulate the maintenance of normal muscle performance and aging in mice. However, the function of MTMR14 in mouse embryonic fibroblasts (MEFs) remains largely unknown. In this study, we established MTMR14 WT and KO MEFs and showed that MTMR14 is localized in whole MEFs, with higher level in nucleus and lower in cytoplasm, partially overlapping with mitochondrial. Compared with the WT control, MTMR14 KO MEFs exhibit a higher proliferation rate and more obvious autophagy. Furthermore, we demonstrate that KO of MTMR14 significantly decreased the mRNA levels of p21 and p27, while increased those of cyclinD and cyclinE. Upon (insulin-like growth factor) IGF stimulation, we also found KO of MTMR14 enhanced the phosphorylation levels of AKT and ERK in MEFs. Based on these findings, we propose that defect of MTMR14 promotes autophagy and cell proliferation in MEFs.


Subject(s)
Autophagy/genetics , Cell Proliferation/genetics , Embryo, Mammalian/metabolism , Phosphoric Monoester Hydrolases/physiology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Phosphoric Monoester Hydrolases/genetics , Polymerase Chain Reaction
13.
Curr Opin Hematol ; 20(4): 289-94, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23615055

ABSTRACT

PURPOSE OF REVIEW: Hematopoietic stem cells (HSCs) residing in the hypoxic niches can both self-renew and give rise to progeny. Multiple regulatory mechanisms for these cellular processes have been identified. Emerging evidence has revealed that metabolism and bioenergetics play important roles in determining stem cell fate in concert with other regulatory networks. In this review, we will discuss recent advances in this field. RECENT FINDINGS: Recent studies have helped define and redefine metabolic regulation of HSCs. Resting quiescent stem cells use primarily anaerobic glycolysis for energy production and this metabolic program is required to maintain a functional quiescent state. However, when they exit this state and rapidly proliferate and differentiate into different blood cell types, a robust up-regulation of energy metabolism is expected to meet the quickly rising energy demand. Dysregulation of metabolism in HSCs results in various blood disorders, including leukemia. SUMMARY: Energy metabolism and HSC activity influence and interlink each other in a highly sophisticated and orchestrated manner. Understanding metabolic regulation of HSC function has significant implications for HSC-based therapies and leukemogenesis research.


Subject(s)
Energy Metabolism/physiology , Hematopoietic Stem Cells/physiology , Cell Differentiation/physiology , Cell Proliferation , Humans , Signal Transduction/physiology
14.
Nat Commun ; 15(1): 5629, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965223

ABSTRACT

Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.


Subject(s)
Epigenesis, Genetic , Histones , Homeostasis , Mice, Knockout , Prostatic Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Receptors, Androgen , Animals , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Histones/metabolism , Male , Humans , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Phosphorylation , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Signal Transduction , Chromatin/metabolism
15.
Sci Adv ; 9(49): eadf9522, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38055827

ABSTRACT

Mitochondria use different substrates for energy production and intermediatory metabolism according to the availability of nutrients and oxygen levels. The role of mitochondrial metabolic flexibility for CD8+ T cell immune response is poorly understood. Here, we report that the deletion or pharmacological inhibition of protein tyrosine phosphatase, mitochondrial 1 (PTPMT1) significantly decreased CD8+ effector T cell development and clonal expansion. In addition, PTPMT1 deletion impaired stem-like CD8+ T cell maintenance and accelerated CD8+ T cell exhaustion/dysfunction, leading to aggravated tumor growth. Mechanistically, the loss of PTPMT1 critically altered mitochondrial fuel selection-the utilization of pyruvate, a major mitochondrial substrate derived from glucose-was inhibited, whereas fatty acid utilization was enhanced. Persistent mitochondrial substrate shift and metabolic inflexibility induced oxidative stress, DNA damage, and apoptosis in PTPMT1 knockout cells. Collectively, this study reveals an important role of PTPMT1 in facilitating mitochondrial utilization of carbohydrates and that mitochondrial flexibility in energy source selection is critical for CD8+ T cell antitumor immunity.


Subject(s)
Mitochondria , PTEN Phosphohydrolase , PTEN Phosphohydrolase/metabolism , Mitochondria/metabolism , Apoptosis , Cell Differentiation , CD8-Positive T-Lymphocytes/metabolism
16.
Elife ; 122023 09 06.
Article in English | MEDLINE | ID: mdl-37672386

ABSTRACT

While mitochondria in different tissues have distinct preferences for energy sources, they are flexible in utilizing competing substrates for metabolism according to physiological and nutritional circumstances. However, the regulatory mechanisms and significance of metabolic flexibility are not completely understood. Here, we report that the deletion of Ptpmt1, a mitochondria-based phosphatase, critically alters mitochondrial fuel selection - the utilization of pyruvate, a key mitochondrial substrate derived from glucose (the major simple carbohydrate), is inhibited, whereas the fatty acid utilization is enhanced. Ptpmt1 knockout does not impact the development of the skeletal muscle or heart. However, the metabolic inflexibility ultimately leads to muscular atrophy, heart failure, and sudden death. Mechanistic analyses reveal that the prolonged substrate shift from carbohydrates to lipids causes oxidative stress and mitochondrial destruction, which in turn results in marked accumulation of lipids and profound damage in the knockout muscle cells and cardiomyocytes. Interestingly, Ptpmt1 deletion from the liver or adipose tissue does not generate any local or systemic defects. These findings suggest that Ptpmt1 plays an important role in maintaining mitochondrial flexibility and that their balanced utilization of carbohydrates and lipids is essential for both the skeletal muscle and the heart despite the two tissues having different preferred energy sources.


Cells are powered by mitochondria, a group of organelles that produce chemical energy in the form of molecules called ATP. This energy is derived from the breakdown of carbohydrates, fats, and proteins. The number of mitochondria in a cell and the energy source they use to produce ATP varies depending on the type of cell. Mitochondria can also switch the molecules they use to produce energy when the cell is responding to stress or disease. The heart and the skeletal muscles ­ which allow movement ­ are two tissues that require large amounts of energy, but it remained unknown whether disrupting mitochondrial fuel selection affects how these tissues work. To answer these questions, Zheng, Li, Li et al. investigated the role of an enzyme found in mitochondria called Ptpmt1. Genetically deleting Ptpmt1 in the heart and skeletal muscle of mice showed that while the development of these organs was not affected, mitochondria in these cells switched from using carbohydrates to using fats as an energy source. Over time, this shift damaged both the mitochondria and the tissues, leading to muscle wasting, heart failure, and sudden death in the mice. This suggests that balanced use of carbohydrates and fats is essential for the muscles and heart. These findings imply that long-term use of medications that alter the fuel that mitochondria use may be detrimental to patients' health and could cause heart dysfunction. This may be important for future drug development, as well as informing decisions about medication taken in the clinic.


Subject(s)
Heart Failure , Animals , Mice , Fatty Acids , Glucose , Heart Failure/genetics , Mice, Knockout , Mitochondria , Muscular Atrophy
17.
Stem Cell Res Ther ; 14(1): 322, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37941041

ABSTRACT

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.


Subject(s)
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
18.
Blood ; 116(18): 3611-21, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20651068

ABSTRACT

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.


Subject(s)
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
19.
Sci Rep ; 12(1): 21832, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36528691

ABSTRACT

Amino acid-mediated metabolism is one of the key catabolic and anabolic processes involved in diverse cellular functions. However, the role of the semi-essential amino acid arginine in normal and malignant hematopoietic cell development is poorly understood. Here we report that a continuous supply of exogenous arginine is required for the maintenance/function of normal hematopoietic stem cells (HSCs). Surprisingly, knockout of Slc7a3 (CAT3), a major L-arginine transporter, does not affect HSCs in steady-state or under stress. Although Slc7a3 is highly expressed in naïve and activated CD8 T cells, neither T cell development nor activation/proliferation is impacted by Slc7a3 depletion. Furthermore, the Slc7a3 deletion does not attenuate leukemia development driven by Pten loss or the oncogenic Ptpn11E76K mutation. Arginine uptake assays reveal that L-arginine uptake is not disrupted in Slc7a3 knockout cells. These data suggest that extracellular arginine is critically important for HSCs, but CAT3 is dispensable for normal hematopoiesis and leukemogenesis.


Subject(s)
Hematopoiesis , Animals , Mice , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Basic/metabolism , Arginine/metabolism , Biological Transport , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Membrane Transport Proteins/metabolism
20.
Blood Adv ; 6(1): 200-206, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34555844

ABSTRACT

Development of normal blood cells is often suppressed in juvenile myelomonocytic leukemia (JMML), a myeloproliferative neoplasm (MPN) of childhood, causing complications and impacting therapeutic outcomes. However, the mechanism underlying this phenomenon remains uncharacterized. To address this question, we induced the most common mutation identified in JMML (Ptpn11E76K) specifically in the myeloid lineage with hematopoietic stem cells (HSCs) spared. These mice uniformly developed a JMML-like MPN. Importantly, HSCs in the same bone marrow (BM) microenvironment were aberrantly activated and differentiated at the expense of self-renewal. As a result, HSCs lost quiescence and became exhausted. A similar result was observed in wild-type (WT) donor HSCs when co-transplanted with Ptpn11E76K/+ BM cells into WT mice. Co-culture testing demonstrated that JMML/MPN cells robustly accelerated differentiation in mouse and human normal hematopoietic stem/progenitor cells. Cytokine profiling revealed that Ptpn11E76K/+ MPN cells produced excessive IL-1ß, but not IL-6, T NF-α, IFN-γ, IL-1α, or other inflammatory cytokines. Depletion of the IL-1ß receptor effectively restored HSC quiescence, normalized their pool size, and rescued them from exhaustion in Ptpn11E76K/+/IL-1R-/- double mutant mice. These findings suggest IL-1ß signaling as a potential therapeutic target for preserving normal hematopoietic development in JMML.


Subject(s)
Hematopoietic Stem Cells , Inflammation , Interleukin-1beta , Leukemia, Myelomonocytic, Juvenile , Animals , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Leukemia, Myelomonocytic, Juvenile/immunology , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Mice , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Receptors, Interleukin-1/deficiency , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL