Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Cell Death Dis ; 15(4): 278, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38637559

Myelodysplastic syndromes (MDS) are a heterogeneous group of pre-leukemic hematopoietic disorders characterized by cytopenia in peripheral blood due to ineffective hematopoiesis and normo- or hypercellularity and morphologic dysplasia in bone marrow (BM). An inflammatory BM microenvironment and programmed cell death of hematopoietic stem/progenitor cells (HSPCs) are thought to be the major causes of ineffective hematopoiesis in MDS. Pyroptosis, apoptosis and necroptosis (collectively, PANoptosis) are observed in BM tissues of MDS patients, suggesting an important role of PANoptosis in MDS pathogenesis. Caspase 8 (Casp8) is a master regulator of PANoptosis, which is downregulated in HSPCs from most MDS patients and abnormally spliced in HSPCs from MDS patients with SRSF2 mutation. To study the role of PANoptosis in hematopoiesis, we generated inducible Casp8 knockout mice (Casp8-/-). Mx1-Cre-Casp8-/- mice died of BM failure within 10 days of polyI:C injections due to depletion of HSPCs. Rosa-ERT2Cre-Casp8-/- mice are healthy without significant changes in BM hematopoiesis within the first 1.5 months after Casp8 deletion. Such mice developed BM failure upon infection or low dose polyI:C/LPS injections due to the hypersensitivity of Casp8-/- HSPCs to infection or inflammation-induced necroptosis which can be prevented by Ripk3 deletion. However, impaired self-renewal capacity of Casp8-/- HSPCs cannot be rescued by Ripk3 deletion due to activation of Ripk1-Tbk1 signaling. Most importantly, mice transplanted with Casp8-/- BM cells developed MDS-like disease within 4 months of transplantation as demonstrated by anemia, thrombocytopenia and myelodysplasia. Our study suggests an essential role for a balance in Casp8, Ripk3-Mlkl and Ripk1-Tbk1 activities in the regulation of survival and self-renewal of HSPCs, the disruption of which induces inflammation and BM failure, resulting in MDS-like disease.


Myelodysplastic Syndromes , Animals , Humans , Mice , Bone Marrow Failure Disorders/complications , Caspase 8/genetics , Caspase 8/metabolism , Inflammation/metabolism , Mice, Knockout , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism
2.
Stem Cell Reports ; 19(1): 100-111, 2024 01 09.
Article En | MEDLINE | ID: mdl-38101400

Lymphoid-primed multipotent progenitor (LMPP)-like and granulocyte-monocyte progenitor (GMP)-like leukemia stem cells (LSCs) co-exist in the blood of most patients with acute myeloid leukemia (AML). Complete elimination of both types of LSCs is required to cure AML. Using an MLL-AF9-induced murine AML model, we studied the role of hematopoietic cytokines in the survival of LMPP- and GMP-like LSCs. We found that SCF or FLT3L promotes the survival of LMPP-like LSCs by stimulating Stat5-mediated Mcl1 expression, whereas interleukin-3 (IL-3) or IL-6 induces the survival of GMP-like LSCs by stimulating Stat3/nuclear factor κB (NF-κB)-mediated Bcl2 expression. Functional study demonstrated that, compared to AML cells cultured in IL-3 and IL-6 medium, AML cells in SCF- or Flt3L-only culture are highly clonogenic in in vitro culture and are highly leukemogenic in vivo. Our study suggests that co-inhibition of both STAT5-MCL1 and STAT3/NF-κB-BCL2 signaling might represent an improved treatment strategy against AML, specifically AML cases with a monocytic phenotype and/or FLT3 mutations.


Interleukin-3 , Leukemia, Myeloid, Acute , Mice , Humans , Animals , Interleukin-3/metabolism , STAT5 Transcription Factor/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NF-kappa B/metabolism , Interleukin-6/metabolism , Leukemia, Myeloid, Acute/genetics , Hematopoietic Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism
3.
J Exp Clin Cancer Res ; 41(1): 294, 2022 Oct 07.
Article En | MEDLINE | ID: mdl-36203205

The family of ten-eleven translocation dioxygenases (TETs) consists of TET1, TET2, and TET3. Although all TETs are expressed in hematopoietic tissues, only TET2 is commonly found to be mutated in age-related clonal hematopoiesis and hematopoietic malignancies. TET2 mutation causes abnormal epigenetic landscape changes and results in multiple stages of lineage commitment/differentiation defects as well as genetic instability in hematopoietic stem/progenitor cells (HSPCs). TET2 mutations are founder mutations (first hits) in approximately 40-50% of cases of TET2-mutant (TET2MT) hematopoietic malignancies and are later hits in the remaining cases. In both situations, TET2MT collaborates with co-occurring mutations to promote malignant transformation. In TET2MT tumor cells, TET1 and TET3 partially compensate for TET2 activity and contribute to the pathogenesis of TET2MT hematopoietic malignancies. Here we summarize the most recent research on TETs in regulating of both normal and pathogenic hematopoiesis. We review the concomitant mutations and aberrant signals in TET2MT malignancies. We also discuss the molecular mechanisms by which concomitant mutations and aberrant signals determine lineage commitment in HSPCs and the identity of hematopoietic malignancies. Finally, we discuss potential strategies to treat TET2MT hematopoietic malignancies, including reverting the methylation state of TET2 target genes and targeting the concomitant mutations and aberrant signals.


Dioxygenases , Hematologic Neoplasms , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Hematologic Neoplasms/genetics , Hematopoiesis/genetics , Humans , Mixed Function Oxygenases , Mutation , Proto-Oncogene Proteins/genetics
4.
Clin Case Rep ; 10(8): e6009, 2022 Aug.
Article En | MEDLINE | ID: mdl-35949412

Acute myeloid leukemia (AML) arises from clonal expansion of malignant hematopoietic precursor cells in the bone marrow. In rare instances, AML can recur with prominent extramedullary manifestations (i.e., leukemia cutis or myeloid sarcoma), either simultaneously or preceding marrow involvement, or as a sole site of primary disease relapse.

6.
Front Med (Lausanne) ; 6: 260, 2019.
Article En | MEDLINE | ID: mdl-31824951

Burn injury has been shown to significantly dampen erythropoiesis in both burn patients and in murine models. Our previous findings elucidated the erythropoietin independent defects in red cell development stages involving erythroid progenitor production and late stage erythroblast enucleation processes. We hypothesized that macrophages (MØ) in erythroblast islands (EBI) could be yet another roadblock impeding erythropoiesis following burn injury. Here we highlight that the methodology to study EBI can be achieved with single cell suspensions using a simple technique such as flow cytometry, as obtaining the central erythroblast island macrophages (EBIMØs) of interest is a delicate process. We elucidated the requisite of EBIMØ from the erythroblast as well as the MØ perspective. In addition to the primary erythropoiesis organ, the bone marrow (BM), spleens were also examined for extra-medullary erythropoiesis. Femurs and spleens were harvested from adult mice (B6D2F1) subjected to 15% total body surface area (TBSA) scald burn (B) or sham burn (S). Total bone marrow cells (TBM) and splenocytes were probed for total erythrons, early and late erythroblasts and EBIMØ by flow cytometry. There was only a marginal increase in the number of EBIMØ after burn, but, between the signatures of EBIMØ, Siglec-1 expression (MFI) was reduced by 40% in B with and a parallel 44% decrease in TBM erythrons in the BM. There were more (2.5-fold) EEBs and less LEBs (2.4-fold) per million TBM cells in B; with a corresponding decrease in Siglec-1 and Ly6G expressions in EBIMØ associated with EEB. Conversely, extra-medullary erythropoiesis was robust in spleens from B. Not only were the numbers of EBIMØs increased in B (p < 0.002), both EEBs and LEBs associated with EBIMØ were higher by 30 and 75%, respectively. Importantly, an increase in Siglec-1 and Vcam1 expressing F480+ splenic macrophages was observed after burn injury. Therefore, stagnant EEBs in the BM after burn injury could be due to low Siglec1 expressing EBIMØ, which perhaps impede their maturation into LEBs and reticulocytes. Repercussion of myeloid cell phenotype specific to BM after burn injury could plausibly account for a defective late stage RBC maturation resulting in anemia of critical illness. Summary Sentence: Characterization of erythroblast island macrophages (EBIMØ) in the bone marrow and spleen at different stages of erythropoiesis after burn injury.

7.
J Cutan Pathol ; 46(9): 653-658, 2019 Sep.
Article En | MEDLINE | ID: mdl-30989699

BACKGROUND: Alopecia areata (AA) is believed to have an autoimmune mechanism in which the hair follicles are targeted by CD4+ and CD8+ lymphocytes. Studies investigating the autoimmune mechanism of other cutaneous diseases, including vitiligo, showed that Treg is a component of cutaneous immune privilege. Our study uses immunohistochemical staining in formalin-fixed, paraffin-embedded tissue to examine the percentage of CD4+ FoxP3+ , CD25+ FoxP3+ , and CD8+ FoxP3+ Treg in AA in human specimens. METHODS: Immunohistochemical double staining for CD4+ FoxP3+ , CD25+ FoxP3+ , and CD8+ FoxP3+ was performed on 12 AA cases and 12 other autoimmune and non-autoimmune cutaneous diseases. The frequency of CD4+ FoxP3+ , CD25+ FoxP3+ , and CD8+ FoxP3+ Treg was counted and expressed as a percentage of total CD4+ , CD25+ , and CD8+ lymphocytes, respectively, in order to account for intersample inflammatory response variability. RESULTS: There was a significant reduction in the mean frequency of CD4+ FoxP3+ and CD25+ FoxP3+ in AA when compared to other autoimmune and non-autoimmune cutaneous diseases. CONCLUSION: Treg is significantly lower in AA when compared to other cutaneous diseases. Additionally, this immunohistochemical-staining protocol may be useful to evaluate Treg in formalin-fixed, paraffin-embedded specimens for other cutaneous diseases. Studies examining Treg in AA and other cutaneous diseases may have implications for future interventions.


Alopecia Areata/immunology , Autoimmune Diseases/immunology , Hair Follicle/immunology , T-Lymphocytes, Regulatory/immunology , Alopecia Areata/pathology , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Female , Hair Follicle/pathology , Humans , Male , T-Lymphocytes, Regulatory/pathology
8.
Brain ; 142(2): 312-321, 2019 02 01.
Article En | MEDLINE | ID: mdl-30649233

There is increasing appreciation for the role of the neurovascular unit in neurodegenerative diseases. We showed previously that the angiogenic and neurotrophic cytokine, vascular endothelial growth factor (VEGF), is suppressed to abnormally low levels in spinocerebellar ataxia type 1 (SCA1), and that replenishing VEGF reverses the cerebellar pathology in SCA1 mice. In that study, however, we used a recombinant VEGF, which is extremely costly to manufacture and biologically unstable as well as immunogenic. To develop a more viable therapy, here we test a synthetic VEGF peptide amphiphile that self-assembles into nanoparticles. We show that this nano-VEGF has potent neurotrophic and angiogenic properties, is well-tolerated, and leads to functional improvement in SCA1 mice even when administered at advanced stages of the disease. This approach can be generalized to other neurotrophic factors or molecules that act in a paracrine manner, offering a novel therapeutic strategy for neurodegenerative conditions.


Nanoparticles/administration & dosage , Spinocerebellar Ataxias/drug therapy , Vascular Endothelial Growth Factor A/administration & dosage , Adult , Animals , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Middle Aged , Nanoparticles/chemistry , Organ Culture Techniques , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Vascular Endothelial Growth Factor A/chemical synthesis
10.
Oncotarget ; 8(5): 8420-8435, 2017 Jan 31.
Article En | MEDLINE | ID: mdl-28039479

We previously reported that autocrine TNF-α (TNF) is responsible for JNK pathway activation in a subset of acute myeloid leukemia (AML) patient samples, providing a survival/proliferation signaling parallel to NF-κB in AML stem cells (LSCs). In this study, we report that most TNF-expressing AML cells (LCs) also express another pro-inflammatory cytokine, IL1ß, which acts in a parallel manner. TNF was produced primarily by LSCs and leukemic progenitors (LPs), whereas IL1ß was mainly produced by partially differentiated leukemic blasts (LBs). IL1ß also stimulates an NF-κB-independent pro-survival and proliferation signal through activation of the JNK pathway. We determined that co-inhibition of signaling stimulated by both TNF and IL1ß synergizes with NF-κB inhibition in eliminating LSCs both ex vivo and in vivo. Our studies show that such treatments are most effective in M4/5 subtypes of AML.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Etanercept/pharmacology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , NF-kappa B/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Nitriles/pharmacology , Sulfones/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Interleukin-1beta/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
11.
Haematologica ; 102(2): 295-307, 2017 02.
Article En | MEDLINE | ID: mdl-27634200

Acquired aplastic anemia is an autoimmune-mediated bone marrow failure syndrome. The mechanism by which such an autoimmune reaction is initiated is unknown. Whether and how the genetic lesions detected in patients cause autoimmune bone marrow failure have not yet been determined. We found that mice with spontaneous deletion of the TGFß-activated kinase-1 gene in a small subset of hematopoietic cells developed bone marrow failure which resembled the clinical manifestations of acquired aplastic anemia patients. Bone marrow failure in such mice could be reversed by depletion of CD4+ T lymphocytes or blocked by knockout of interferon-γ, suggesting a Th1-cell-mediated autoimmune mechanism. The onset and progression of bone marrow failure in such mice were significantly accelerated by the inactivation of tumor necrosis factor-α signaling. Tumor necrosis factor-α restricts autoimmune bone marrow failure by inhibiting type-1 T-cell responses and maintaining the function of myeloid-derived suppressor cells. Furthermore, we determined that necroptosis among a small subset of mutant hematopoietic cells is the cause of autoimmune bone marrow failure because such bone marrow failure can be prevented by deletion of receptor interacting protein kinase-3 Our study suggests a novel mechanism to explain the pathogenesis of autoimmune bone marrow failure.


Apoptosis , Autoimmunity , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Mutation , Necrosis , Anemia, Aplastic/etiology , Anemia, Aplastic/metabolism , Anemia, Aplastic/mortality , Anemia, Aplastic/pathology , Animals , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Bone Marrow/pathology , Disease Models, Animal , Disease Progression , Female , Hematopoiesis/genetics , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Interferon-gamma/deficiency , Lymphocyte Activation , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Knockout , Necrosis/genetics , Necrosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Leuk Res Rep ; 6: 27-28, 2016.
Article En | MEDLINE | ID: mdl-27924280

The exon 14 JAK2 V617F mutation has been well established as a driver mutation in polycythemia vera (PV) and other myeloproliferative neoplasms. JAK2 exon 12 mutations have also been implicated in PV, although patients with these mutations may show isolated erythrocytosis. Recently additional JAK2point mutations have been described-all in regions encoding the pseudokinase domain that regulates the tyrosine kinase activity of JAK2. We present a case of a patient with erythrocytosis and an exon 13 G571S mutation, and discuss the putative role of this mutation in myeloproliferative neoplasms.

13.
Cell Rep ; 10(12): 2055-68, 2015 Mar 31.
Article En | MEDLINE | ID: mdl-25801032

Mutations and inactivation of phosphatase and tensin homolog deleted from chromosome 10 (PTEN) are observed in 15%-25% of cases of human T cell acute lymphoblastic leukemia (T-ALL). Pten deletion induces myeloproliferative disorders (MPDs), acute myeloid leukemia (AML), and/or T-ALL in mice. Previous studies attributed Pten-loss-related hematopoietic defects and leukemogenesis to excessive activation of phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling. Although inhibition of this signal dramatically suppresses the growth of PTEN-null T-ALL cells in vitro, treatment with inhibitors of this pathway does not cause a complete remission in vivo. Here, we report that focal adhesion kinase (Fak), a protein substrate of Pten, also contributes to T-ALL development in Pten-null mice. Inactivation of the FAK signaling pathway by either genetic or pharmacologic methods significantly sensitizes both murine and human PTEN-null T-ALL cells to PI3K/AKT/mTOR inhibition when cultured in vitro on feeder layer cells or a matrix and in vivo.


Focal Adhesion Protein-Tyrosine Kinases/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Humans , Leukemia/metabolism , Mice , Mice, Knockout , Signal Transduction/drug effects
14.
Diagn Cytopathol ; 43(1): 60-5, 2015 Jan.
Article En | MEDLINE | ID: mdl-24623571

T cell-derived malignant lymphoma is rarely detected as a bladder neoplasm. A literature review for anaplastic large cell lymphoma (ALCL) involving urinary bladder reveals only seven previously reported cases. Here, we report a case of a 59-year-old HIV-negative man with ALK-positive ALCL. He presented an unusual clinical course with initial consideration of adult onset Still's Disease (AOSD) due to his negative results searching for malignancy and infectious diseases. He rapidly developed macrophage activation (hemophagocytic) syndrome and experienced an unusual rapid disease progression and died in 39 days after onset of symptoms. Compared to previously reported cases, the current case of ALK-1-positive ALCL is a rare case with an unusual presentation. From this case, we learned that ALCL is one malignancy that should be considered and screened in patients with suspected AOSD. Also, T-cell lymphoma associated hemophagocytic syndrome should be considered in a patient with sustained corticosteroid-resistant spike fever, high serum ferritin, and rapid exacerbation of the disease course.


Lymphoma, Large-Cell, Anaplastic/pathology , Urinary Bladder/pathology , Activin Receptors, Type II/genetics , Diagnosis, Differential , Humans , Lymphoma, Large-Cell, Anaplastic/diagnosis , Lymphoma, Large-Cell, Anaplastic/genetics , Male , Middle Aged , Still's Disease, Adult-Onset/diagnosis
17.
J Pediatr Hematol Oncol ; 36(2): e125-7, 2014 Mar.
Article En | MEDLINE | ID: mdl-23689288

BACKGROUND: Burkitt lymphoma (BL) is an aggressive, rapidly proliferating neoplasm of B-cell origin. A late recurrence should be investigated to differentiate whether it is a true relapse or a de novo lymphoma as this has therapeutic implications. OBSERVATIONS: We report an HIV-negative white male individual, who at the age of 14 presented with recurrent BL in the abdomen occurring 6 years after successful treatment. Analysis of VDJ rearrangement showed marked dissimilarity in clonal peaks between the 2 tumors, suggesting that each tumor was associated with separate origins. The second tumor was treated as a de novo BL, and the patient remained in complete remission 2 years from recurrence without any evidence of the disease. CONCLUSION: We present a case with 2 distinct BLs verified by the VDJ rearrangement analysis in a non-HIV infected individual. Our case supports the finding that a genetically discrete BL can be treated as if it were a new tumor, thereby reducing chemotherapeutic burden and treatment-related morbidity associated with the regimens for relapsed or refractory disease. Analysis of VDJ rearrangement seems to be a reliable assessment of tumor clonality.


Burkitt Lymphoma/genetics , Immunoglobulin Heavy Chains/genetics , Neoplasms, Second Primary/genetics , V(D)J Recombination/genetics , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Burkitt Lymphoma/diagnosis , Burkitt Lymphoma/drug therapy , Flow Cytometry , Humans , Male , Neoplasms, Second Primary/diagnosis , Neoplasms, Second Primary/drug therapy , Polymerase Chain Reaction
18.
Am J Clin Pathol ; 140(5): 686-92, 2013 Nov.
Article En | MEDLINE | ID: mdl-24124148

OBJECTIVES: To examine the utility of CD11c expression on monocytes in normal controls and patients with chronic myelomonocytic leukemia (CMML) (n = 23) with flow cytometric immunophenotyping. METHODS: Twenty-three CMML samples and 10 control bone marrows submitted for lymphoma staging without evidence of disease were examined. RESULTS: Monocytes in CMML samples ranged from 4% to 35%. Expression of at least one aberrant monocytic marker was found on the monocytes in 18 (82%) of 22 evaluable cases. The most common aberrancy was underexpression of CD11c (n = 15), while none of the bone marrow controls showed underexpression of CD11c. CONCLUSIONS: A distinct heterogeneous population of monocytic cells with underexpression of CD11c was identified in all these cases. CD11c underexpression was independent of other aberrancies, including HLA-DR underexpression (n = 14), aberrant CD56 expression (n = 11), and underexpression of CD33, CD38, and CD14 (n = 6, 5, and 5, respectively), supporting the utility of CD11c expression status on monocytes in establishing a CMML diagnosis.


CD11c Antigen/blood , Leukemia, Myelomonocytic, Chronic/pathology , Monocytes/pathology , Antigens, CD/blood , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Female , HLA-DR Antigens/blood , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelomonocytic, Chronic/metabolism , Leukocytosis/complications , Leukocytosis/metabolism , Leukocytosis/pathology , Male , Monocytes/metabolism , Neoplasm Staging
19.
J Trauma ; 71(5): 1288-96, 2011 Nov.
Article En | MEDLINE | ID: mdl-22071930

BACKGROUND: Anemia in burn patients is due to surgical blood loss and anemia of critical illness. Because the commitment paradigm of common bone marrow progenitors dictates the production of erythroid, myeloid, and lymphoid cells, we hypothesized that skewed bone marrow lineage commitment decreases red cell production and causes anemia after a burn injury. METHODS: After anesthesia, B(6)D(2)F(1) mice received a 15% total body surface area dorsal scald burn. The sham group did not receive scald burn. Femoral bone marrow was harvested on 2, 5, 7, 14, and 21 postburn days (PBD). Total bone marrow cells were labeled with specific antibodies to erythroid (CD71/Ter119), myeloid (CD11b), and lymphoid (CD19) lineages and analyzed by flow cytometry. To test whether erythropoietin (EPO) could increase red blood cell production, EPO was administered to sham and burn animals and their reticulocyte response was measured on PBD 2 and PBD 7. RESULTS: Burn injury reduced the erythroid cells of the bone marrow from 35% in sham to 17% by PBD 5 and remained at similar level until PBD 21. Myeloid cells, however, increased from 42% in sham to 60% on PBD 5 and 77% on PBD 21. Burn injury reduced reticulocyte counts on PBD 2 and PBD 7 indicating that the erythroid compartment is severely depleted. This depleted compartment, however, responded to EPO but was not sufficient to change red cell production. CONCLUSION: Burn injury skews the bone marrow hematopoietic commitment away from erythroid and toward myeloid cells. Shrinkage of the erythroid compartment contributes to resistance to EPO and the anemia of critical illness.


Anemia/etiology , Anemia/metabolism , Bone Marrow Cells/metabolism , Burns/complications , Erythroid Cells/metabolism , Hematopoietic System/metabolism , Lymphocytes/metabolism , Myeloid Cells/metabolism , Analysis of Variance , Animals , Flow Cytometry , Male , Mice , Random Allocation , Reticulocytes/metabolism
20.
Nat Med ; 17(11): 1445-7, 2011 Oct 16.
Article En | MEDLINE | ID: mdl-22001907

Spinocerebellar ataxia type 1 (SCA1) is an adult-onset, dominantly inherited neurodegenerative disease caused by expansion of a glutamine repeat tract in ataxin-1 (ATXN1). Although the precise function of ATXN1 remains elusive, it seems to be involved in transcriptional repression. We find that mutant ATXN1 represses transcription of the neurotrophic and angiogenic factor vascular endothelial growth factor (VEGF). Genetic overexpression or pharmacologic infusion of recombinant VEGF mitigates SCA1 pathogenesis, suggesting a new therapeutic strategy for this disease.


Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Ataxin-1 , Ataxins , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Spinocerebellar Ataxias/pathology , Vascular Endothelial Growth Factor A/genetics
...