Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Biol Chem ; 300(8): 107542, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992436

ABSTRACT

Diamond Blackfan Anemia (DBA) is a rare macrocytic red blood cell aplasia that usually presents within the first year of life. The vast majority of patients carry a mutation in one of approximately 20 genes that results in ribosomal insufficiency with the most significant clinical manifestations being anemia and a predisposition to cancers. Nemo-like Kinase (NLK) is hyperactivated in the erythroid progenitors of DBA patients and inhibition of this kinase improves erythropoiesis, but how NLK contributes to the pathogenesis of the disease is unknown. Here we report that activated NLK suppresses the critical upregulation of mitochondrial biogenesis required in early erythropoiesis. During normal erythropoiesis, mTORC1 facilitates the translational upregulation of Transcription factor A, mitochondrial (TFAM), and Prohibin 2 (PHB2) to increase mitochondrial biogenesis. In our models of DBA, active NLK phosphorylates the regulatory component of mTORC1, thereby suppressing mTORC1 activity and preventing mTORC1-mediated TFAM and PHB2 upregulation and subsequent mitochondrial biogenesis. Improvement of erythropoiesis that accompanies NLK inhibition is negated when TFAM and PHB2 upregulation is prevented. These data demonstrate that a significant contribution of NLK on the pathogenesis of DBA is through loss of mitochondrial biogenesis.


Subject(s)
Anemia, Diamond-Blackfan , Erythropoiesis , Mechanistic Target of Rapamycin Complex 1 , Mitochondria , Organelle Biogenesis , Prohibitins , Protein Serine-Threonine Kinases , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mitochondria/metabolism , Mitochondria/pathology , Animals , Mice , Repressor Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation , Mitochondrial Proteins
2.
Stem Cells ; 41(6): 560-569, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36987811

ABSTRACT

Diamond Blackfan anemia (DBA) is an inherited bone marrow failure syndrome associated with severe anemia, congenital malformations, and an increased risk of developing cancer. The chromatin-binding special AT-rich sequence-binding protein-1 (SATB1) is downregulated in megakaryocyte/erythroid progenitors (MEPs) in patients and cell models of DBA, leading to a reduction in MEP expansion. Here we demonstrate that SATB1 expression is required for the upregulation of the critical erythroid factors heat shock protein 70 (HSP70) and GATA1 which accompanies MEP differentiation. SATB1 binding to specific sites surrounding the HSP70 genes promotes chromatin loops that are required for the induction of HSP70, which, in turn, promotes GATA1 induction. This demonstrates that SATB1, although gradually downregulated during myelopoiesis, maintains a biological function in early myeloid progenitors.


Subject(s)
Anemia, Diamond-Blackfan , Matrix Attachment Region Binding Proteins , Humans , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Megakaryocytes/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Cell Differentiation/genetics , Transcription Factors/metabolism , Anemia, Diamond-Blackfan/metabolism , Chromatin/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism
3.
J Biol Chem ; 297(3): 100988, 2021 09.
Article in English | MEDLINE | ID: mdl-34298020

ABSTRACT

Nemo-like kinase (NLK) is a member of the mitogen-activated protein kinase family of kinases and shares a highly conserved kinase domain with other mitogen-activated protein kinase family members. The activation of NLK contributes to the pathogenesis of Diamond-Blackfan anemia (DBA), reducing c-myb expression and mechanistic target of rapamycin activity, and is therefore a potential therapeutic target. Unlike other anemias, the hematopoietic effects of DBA are largely restricted to the erythroid lineage. Mutations in ribosomal genes induce ribosomal insufficiency and reduced protein translation, dramatically impacting early erythropoiesis in the bone marrow of patients with DBA. We sought to identify compounds that suppress NLK and increases erythropoiesis in ribosomal insufficiency. We report that the active component of ginseng, ginsenoside Rb1, suppresses NLK expression and improves erythropoiesis in in vitro models of DBA. Ginsenoside Rb1-mediated suppression of NLK occurs through the upregulation of miR-208, which binds to the 3'-UTR of NLK mRNA and targets it for degradation. We also compare ginsenoside Rb1-mediated upregulation of miR-208 with metformin-mediated upregulation of miR-26. We conclude that targeting NLK expression through miRNA binding of the unique 3'-UTR is a viable alternative to the challenges of developing small-molecule inhibitors to target the highly conserved kinase domain of this specific kinase.


Subject(s)
Anemia, Diamond-Blackfan/pathology , Erythropoiesis/drug effects , Ginsenosides/pharmacology , Panax/chemistry , Protein Serine-Threonine Kinases/drug effects , 3' Untranslated Regions , Animals , Humans
4.
Curr Opin Hematol ; 24(4): 307-313, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28306669

ABSTRACT

PURPOSE OF REVIEW: SH2 domain-containing tyrosine phosphatase 2 (SHP2), encoded by PTPN11 plays an important role in regulating signaling from cell surface receptor tyrosine kinases during normal development as well as oncogenesis. Herein we review recently discovered roles of SHP2 in normal and aberrant hematopoiesis along with novel strategies to target it. RECENT FINDINGS: Cell autonomous role of SHP2 in normal hematopoiesis and leukemogenesis has long been recognized. The review will discuss the newly discovered role of SHP2 in lineage specific differentiation. Recently, a noncell autonomous role of oncogenic SHP2 has been reported in which activated SHP2 was shown to alter the bone marrow microenvironment resulting in transformation of donor derived normal hematopoietic cells and development of myeloid malignancy. From being considered as an 'undruggable' target, recent development of allosteric inhibitor has made it possible to specifically target SHP2 in receptor tyrosine kinase driven malignancies. SUMMARY: SHP2 has emerged as an attractive target for therapeutic targeting in hematological malignancies for its cell autonomous and microenvironmental effects. However a better understanding of the role of SHP2 in different hematopoietic lineages and its crosstalk with signaling pathways activated by other genetic lesions is required before the promise is realized in the clinic.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Hematopoiesis , Leukemia/genetics , Leukemia/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Carrier Proteins , Hematopoiesis/genetics , Humans , Leukemia/drug therapy , Molecular Targeted Therapy , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Signal Transduction/drug effects
5.
Exp Hematol ; 91: 65-77, 2020 11.
Article in English | MEDLINE | ID: mdl-32926965

ABSTRACT

Diamond-Blackfan anemia (DBA) results from haploinsufficiency of ribosomal protein subunits in hematopoietic progenitors in the earliest stages of committed erythropoiesis. Nemo-like kinase (NLK) is chronically hyperactivated in committed erythroid progenitors and precursors in multiple human and murine models of DBA. Inhibition of NLK activity and suppression of NLK expression both improve erythroid expansion in these models. Metformin is a well-tolerated drug for type 2 diabetes with multiple cellular targets. Here we demonstrate that metformin improves erythropoiesis in human and zebrafish models of DBA. Our data indicate that the effects of metformin on erythroid proliferation and differentiation are mediated by suppression of NLK expression through induction of miR-26a, which recognizes a binding site within the NLK 3' untranslated region (3'UTR) to facilitate transcript degradation. We propose that induction of miR-26a is a potentially novel approach to treatment of DBA and could improve anemia in DBA patients without the potentially adverse side effects of metformin in a DBA patient population.


Subject(s)
Anemia, Diamond-Blackfan/drug therapy , Erythropoiesis/drug effects , Hematinics/therapeutic use , Metformin/therapeutic use , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3' Untranslated Regions/genetics , Anemia, Diamond-Blackfan/genetics , Animals , Cells, Cultured , Colony-Forming Units Assay , Disease Models, Animal , Erythropoiesis/genetics , Gene Expression Regulation/drug effects , Genes, Reporter , Hematinics/pharmacology , Humans , Metformin/pharmacology , MicroRNAs/genetics , RNA Stability , RNA, Small Interfering/pharmacology , Recombinant Proteins/metabolism , Species Specificity , Up-Regulation/drug effects , Zebrafish
6.
Indian J Otolaryngol Head Neck Surg ; 71(3): 352-359, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31559203

ABSTRACT

The incorporation of telemedicine and artificial intelligence for early screening and assessment of severity of life-style disorders has a great potential for better assessment in a busy outpatient clinic and thereby curtail down the related morbidities. A computer based algorithm based upon standardized questionnaire (from established assessment tools) is designed to assess the risk of obstructive sleep apnoea syndrome (OSAS). In addition the incorporation of basic screening questions of anamnesis help in suggesting a probable diagnosis of sleep related disorder as well. The overall data at our center has been analyzed to establish the existing pattern of sleep related disorders. Of 850 healthy subjects screened, prevalence of snoring was 20.47% while OSAS was seen in 4.20% (N = 25) in males and 2.64% (N = 8) in females. The parasomnia was most prevalent (14.71%), followed by insomnia (10.24%), periodic leg movement (6.59%), bruxism (1.65%) and narcolepsy (0.59%). Hypertension, laryngopharyngeal reflux and obesity were the common co-morbidities in OSAS while family history of hypertension and diabetes were common in snorers. A significant association with OSA was seen with diabetes mellitus, neck circumference and nasal obstruction, while, obesity and apnoeic episodes were more significantly associated with OSA than snorers. Increased waist to hip ratio was appreciated in both the OSAS and snorers. The algorithm based online assessment is likely to diagnose the occult clinical cases as well as assess the risk of OSAS. In routine outpatient clinic, a clinician may better assess the patient morbidity with a comprehensive availability of symptoms and moreover enhance the post-treatment compliance. In addition a smartphone based computerized assessment for general population may be designed for other lifestyle disorders as well.

SELECTION OF CITATIONS
SEARCH DETAIL