• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 88
  • 77
  • 24
  • 24
  • 7
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 1
  • 1
  • Tagged with
  • 276
  • 96
  • 80
  • 58
  • 53
  • 40
  • 34
  • 31
  • 27
  • 25
  • 22
  • 22
  • 21
  • 20
  • 20
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
271

Efeitos in vitro de soro de pacientes com nefrite lúpica ativa em células de linhagem osteoblástica humana hFOB 1.19 / The in vitro effects of the serum of patients with active lupus nephritis on the human osteoblast-like cell model hFOB 1.19

Lima, Ana Paula Calheiros de 07 December 2018 (has links)
INTRODUÇÃO: Perda óssea é um achado comum em pacientes com Nefrite Lúpica (NL), mesmo naqueles com diagnóstico recente. Algumas evidências indicam um aumento na osteoclastogênese como um dos distúrbios principais no processo de remodelamento ósseo. O objetivo deste estudo foi investigar algumas vias de sinalização (RANKL/OPG, Wnt/Beta-catenin and Th17/IL-17) possivelmente envolvidas na osteoclastogênese anormal detectada em mulheres jovens ao diagnóstico de nefrite lúpica ativa, assim como avaliar a ação da vitamina D (VitD) nesse cenário e sua correlação com fatores inflamatórios. MÉTODOS: Realizamos culturas com a linhagem de células osteoblásticas humanas hFOB 1.19 (ATCC) e as dividimos em um grupo suplementado com soro de pacientes lúpicas (NL) (n=15) e em um grupo com soro de controles saudáveis (CS) (n=15) em vez de soro fetal bovino (SFB). Em seguida, adicionamos 1,25-dihidroxivitamina D3 (1,25(OH)2D3) em dois subgrupos nas concentrações 10-9M e 10-7M, resultando em 6 grupos: CS, CS+vitD 10-9M, CS+vitD 10-7M, NL, NL+vitD 10-9M, NL+vitD 10-7M). Após 48h da adição de 1,25(OH)2D3 ao meio de cultura, células hFOB foram tripsinizadas e separado o lisato celular de cada grupo. Ensaios de citometria de fluxo e multiplex foram realizados para quantificação das seguintes proteínas do lisato cellular: CD166, CD54, fosfatase alcalina, RANKL, OPG, CD14, TLR4, NF-KappaB, SOST, DKK-1, Beta-catenina, IL-1-beta, IL-2, IL-6, TNF-alfa, IL-17A, IL-17F, IL-21 and IL-22. RT-PCR foi empregado para quantificação de mRNA dos genes RANKL, SOST, OPG e Beta-catenina. RESULTADOS: Pacientes com NL evidenciaram maiores níveis séricos de DKK-1 (2802,04 ± 1380,06 x 696,30 ± 421,22pg/ml, p < 0,001), OPG (560,12 ± 333,56 x 340,24 ± 102,08pg/ml, p=0,0212), TNF-alfa (9,63 ± 14,49 x 1,27 ± 0,35pg/ml, p=0,0337), IL-6 (15,58±39,08 x 8,02±3,49, p= 0,0053) and IL-2 (3,36 ± 3,06 x 1,54 ± 0,9pg/ml, p=0,0353) do que CS. Após exposição ao meio enriquecido com soro de pacientes com NL, células hFOB 1.19 apresentaram maior nível de mRNA de RANKL (p=0,045)) e menor nível de proteína OPG (178,81 ± 66,40 x 298,76 ± 114,94pg/mg, p=0,0016). Suplementação com 1,25(OH)2D3 aumentou a diferença da expressão das proteínas DKK-1 (673,03 ± 171,93 x 456,69 ± 234,53pg/mg, p=0,0215), IL-6 (0,80 ± 0,25pg/mg x 0,66 ± 0,18, p=0,0417) and IL-2 (4,97 ± 2,2 x 3,90 ± 1,66pg/mg, p=0,042) entre hFOB NL comparados com hFOB CS, enquanto diminuiu o nível de mRNA de Beta-catenina em células do grupo NL. DISCUSSÃO: Dentro das limitações deste estudo, os resultados sugerem que os maiores níveis séricos de citocinas pró-inflamatórias, como TNF-alfa, IL-6 e talvez IL-2, detectadas em pacientes com NL pode ter induzido a maior expressão osteoblástica de RANKL, representada pelo maior nível de mRNA RANKL em células do grupo NL, e suprimido OPG, conforme a diminuição observada na quantificação proteica de OPG nos lisatos celulares, o que pode ter contribuído para a aumentada osteoclastogênese evidenciada pela biópsia óssea dessas pacientes. A adição de 1,25(OH)2D3 não preveniu os efeitos inflamatórios do soro de pacientes com NL ativa em células hFOB 1.19 neste estudo / INTRODUCTION: Bone loss is a common finding in Lupus Nephritis (LN) patients even in those recently diagnosed. Some evidences indicate an increased osteoclastogenesis as the main disturb of the bone remodeling process. The aim of this study was to investigate some pathways (RANK-L/OPG, Wnt/?-catenin and Th17/IL-17) possibly involved in the abnormal osteoclastogenesis detected in women at the diagnosis of proliferative LN as well as evaluating the action of vitamin D (vitD) in this scenario and their correlation with inflammatory factors. METHODS: We cultured the human osteoblastic cell line hFOB 1.19 (ATCC), and divided cultures into those supplemented with serum from healthy controls (HC) (n=15) and LN patients (n=15) instead of fetal bovine serum (FBS). Then 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] was added in two subgroups at the concentrations of 10-9M e 10-7M while vitD was absent in one subgroup in both HC and LN cultures (HC, HC+vitD 10-9M, HC+vitD 10-7M, LN, LN+vitD 10-9M, LN+vitD 10-7M) . After 48h of vitD addition, hFOB cultures were trypsinized. Flow cytometry and multiplex assays were performed to test CD166, CD54, alkaline phosphatase, RANKL, OPG, CD14, TLR4, NF-KappaB, SOST, DKK-1, ?-catenin, IL-1Beta, IL-2, IL-6, TNF-alfa, IL-17A, IL-17F, IL-21 and IL-22 concentrations in the cell lysates. Polymerase reaction chain (RT-PCR) assays analyzed the expression of RANKL, SOST, OPG and Beta-catenin mRNA. RESULTS: LN patients showed higher serum levels of DKK-1 (2802.04 ± 1380.06 x 696.3 ± 421.22pg/ml, p < 0.001), OPG (560.12 ± 333.56 x 340.24 ± 102.08pg/ml, p=0.0212), TNF-alfa (9.63 ± 14.49 x 1.27 ± 0.35pg/ml, p=0.0337), IL-6 (15.58±39.08 x 8.02±3.49, 0.0053) and IL-2 (3.36 ± 3.06 x 1.54 ± 0.9pg/ml, p=0.0353) than HCs. After exposure to medium enriched with LN serum, osteoblasts expressed higher RANKL mRNA (fold change 1.573, p=0.045) and lower OPG protein (178.81 ± 66.40 x 298.76 ± 114.94pg/mg, p=0.0016). 1,25(OH)2D3 supplementation increased the difference between LN and HC expression of DKK-1 (673.03 ± 171.93 x 456.69 ± 234.53pg/mg, p=0.0215), IL-6 (0.80 ± 0.25pg/mg x 0.66 ± 0.18, p=0.0417) and IL-2 (4.97 ± 2.2 x 3.90 ± 1,66pg/mg, p=0.042) proteins and diminished Beta-catenin mRNA in LN cells. DISCUSSION: Within the limitations of this study, the results suggest that the higher serum levels of proinflammatory cytokines, such as TNF-alfa, IL-6 and perhaps IL-2, detected in LN patients would possibly have induced RANKL genes, as demonstrated by an enhanced RANKL mRNA expression in LN osteoblasts, and suppressed OPG protein in cell lysates, which would have contributed to the increased osteoclastogenesis detected in bone biopsies of women with new onset of LN. 1,25(OH)2D3 addition to osteoblast cultures did not prevent the effects of inflammatory LN serum in vitro
272

Efeitos in vitro de soro de pacientes com nefrite lúpica ativa em células de linhagem osteoblástica humana hFOB 1.19 / The in vitro effects of the serum of patients with active lupus nephritis on the human osteoblast-like cell model hFOB 1.19

Ana Paula Calheiros de Lima 07 December 2018 (has links)
INTRODUÇÃO: Perda óssea é um achado comum em pacientes com Nefrite Lúpica (NL), mesmo naqueles com diagnóstico recente. Algumas evidências indicam um aumento na osteoclastogênese como um dos distúrbios principais no processo de remodelamento ósseo. O objetivo deste estudo foi investigar algumas vias de sinalização (RANKL/OPG, Wnt/Beta-catenin and Th17/IL-17) possivelmente envolvidas na osteoclastogênese anormal detectada em mulheres jovens ao diagnóstico de nefrite lúpica ativa, assim como avaliar a ação da vitamina D (VitD) nesse cenário e sua correlação com fatores inflamatórios. MÉTODOS: Realizamos culturas com a linhagem de células osteoblásticas humanas hFOB 1.19 (ATCC) e as dividimos em um grupo suplementado com soro de pacientes lúpicas (NL) (n=15) e em um grupo com soro de controles saudáveis (CS) (n=15) em vez de soro fetal bovino (SFB). Em seguida, adicionamos 1,25-dihidroxivitamina D3 (1,25(OH)2D3) em dois subgrupos nas concentrações 10-9M e 10-7M, resultando em 6 grupos: CS, CS+vitD 10-9M, CS+vitD 10-7M, NL, NL+vitD 10-9M, NL+vitD 10-7M). Após 48h da adição de 1,25(OH)2D3 ao meio de cultura, células hFOB foram tripsinizadas e separado o lisato celular de cada grupo. Ensaios de citometria de fluxo e multiplex foram realizados para quantificação das seguintes proteínas do lisato cellular: CD166, CD54, fosfatase alcalina, RANKL, OPG, CD14, TLR4, NF-KappaB, SOST, DKK-1, Beta-catenina, IL-1-beta, IL-2, IL-6, TNF-alfa, IL-17A, IL-17F, IL-21 and IL-22. RT-PCR foi empregado para quantificação de mRNA dos genes RANKL, SOST, OPG e Beta-catenina. RESULTADOS: Pacientes com NL evidenciaram maiores níveis séricos de DKK-1 (2802,04 ± 1380,06 x 696,30 ± 421,22pg/ml, p < 0,001), OPG (560,12 ± 333,56 x 340,24 ± 102,08pg/ml, p=0,0212), TNF-alfa (9,63 ± 14,49 x 1,27 ± 0,35pg/ml, p=0,0337), IL-6 (15,58±39,08 x 8,02±3,49, p= 0,0053) and IL-2 (3,36 ± 3,06 x 1,54 ± 0,9pg/ml, p=0,0353) do que CS. Após exposição ao meio enriquecido com soro de pacientes com NL, células hFOB 1.19 apresentaram maior nível de mRNA de RANKL (p=0,045)) e menor nível de proteína OPG (178,81 ± 66,40 x 298,76 ± 114,94pg/mg, p=0,0016). Suplementação com 1,25(OH)2D3 aumentou a diferença da expressão das proteínas DKK-1 (673,03 ± 171,93 x 456,69 ± 234,53pg/mg, p=0,0215), IL-6 (0,80 ± 0,25pg/mg x 0,66 ± 0,18, p=0,0417) and IL-2 (4,97 ± 2,2 x 3,90 ± 1,66pg/mg, p=0,042) entre hFOB NL comparados com hFOB CS, enquanto diminuiu o nível de mRNA de Beta-catenina em células do grupo NL. DISCUSSÃO: Dentro das limitações deste estudo, os resultados sugerem que os maiores níveis séricos de citocinas pró-inflamatórias, como TNF-alfa, IL-6 e talvez IL-2, detectadas em pacientes com NL pode ter induzido a maior expressão osteoblástica de RANKL, representada pelo maior nível de mRNA RANKL em células do grupo NL, e suprimido OPG, conforme a diminuição observada na quantificação proteica de OPG nos lisatos celulares, o que pode ter contribuído para a aumentada osteoclastogênese evidenciada pela biópsia óssea dessas pacientes. A adição de 1,25(OH)2D3 não preveniu os efeitos inflamatórios do soro de pacientes com NL ativa em células hFOB 1.19 neste estudo / INTRODUCTION: Bone loss is a common finding in Lupus Nephritis (LN) patients even in those recently diagnosed. Some evidences indicate an increased osteoclastogenesis as the main disturb of the bone remodeling process. The aim of this study was to investigate some pathways (RANK-L/OPG, Wnt/?-catenin and Th17/IL-17) possibly involved in the abnormal osteoclastogenesis detected in women at the diagnosis of proliferative LN as well as evaluating the action of vitamin D (vitD) in this scenario and their correlation with inflammatory factors. METHODS: We cultured the human osteoblastic cell line hFOB 1.19 (ATCC), and divided cultures into those supplemented with serum from healthy controls (HC) (n=15) and LN patients (n=15) instead of fetal bovine serum (FBS). Then 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] was added in two subgroups at the concentrations of 10-9M e 10-7M while vitD was absent in one subgroup in both HC and LN cultures (HC, HC+vitD 10-9M, HC+vitD 10-7M, LN, LN+vitD 10-9M, LN+vitD 10-7M) . After 48h of vitD addition, hFOB cultures were trypsinized. Flow cytometry and multiplex assays were performed to test CD166, CD54, alkaline phosphatase, RANKL, OPG, CD14, TLR4, NF-KappaB, SOST, DKK-1, ?-catenin, IL-1Beta, IL-2, IL-6, TNF-alfa, IL-17A, IL-17F, IL-21 and IL-22 concentrations in the cell lysates. Polymerase reaction chain (RT-PCR) assays analyzed the expression of RANKL, SOST, OPG and Beta-catenin mRNA. RESULTS: LN patients showed higher serum levels of DKK-1 (2802.04 ± 1380.06 x 696.3 ± 421.22pg/ml, p < 0.001), OPG (560.12 ± 333.56 x 340.24 ± 102.08pg/ml, p=0.0212), TNF-alfa (9.63 ± 14.49 x 1.27 ± 0.35pg/ml, p=0.0337), IL-6 (15.58±39.08 x 8.02±3.49, 0.0053) and IL-2 (3.36 ± 3.06 x 1.54 ± 0.9pg/ml, p=0.0353) than HCs. After exposure to medium enriched with LN serum, osteoblasts expressed higher RANKL mRNA (fold change 1.573, p=0.045) and lower OPG protein (178.81 ± 66.40 x 298.76 ± 114.94pg/mg, p=0.0016). 1,25(OH)2D3 supplementation increased the difference between LN and HC expression of DKK-1 (673.03 ± 171.93 x 456.69 ± 234.53pg/mg, p=0.0215), IL-6 (0.80 ± 0.25pg/mg x 0.66 ± 0.18, p=0.0417) and IL-2 (4.97 ± 2.2 x 3.90 ± 1,66pg/mg, p=0.042) proteins and diminished Beta-catenin mRNA in LN cells. DISCUSSION: Within the limitations of this study, the results suggest that the higher serum levels of proinflammatory cytokines, such as TNF-alfa, IL-6 and perhaps IL-2, detected in LN patients would possibly have induced RANKL genes, as demonstrated by an enhanced RANKL mRNA expression in LN osteoblasts, and suppressed OPG protein in cell lysates, which would have contributed to the increased osteoclastogenesis detected in bone biopsies of women with new onset of LN. 1,25(OH)2D3 addition to osteoblast cultures did not prevent the effects of inflammatory LN serum in vitro
273

Étude des effets du phénotype de sénescence des cellules stromales de la moelle osseuse sur les fonctions hématopoïétiques

Carbonneau, Cynthia 12 1900 (has links)
L’irradiation (IR) est utilisée dans le traitement de plusieurs cancers et désordres hématologiques, en particulier dans les protocoles de conditionnement précédents les transplantations de moelle osseuse. L’emploi de doses réduites d’IR semble favoriser le succès de la prise de greffe. Cette observation soulève un point de plus en plus discuté dans la littérature, soit l’importance de l’intégrité du microenvironnement pour la transplantation et le bon fonctionnement de l’hématopoïèse. L’IR induit la sénescence des cellules stromales de la moelle osseuse in vitro. Ce mécanisme de défense cellulaire entraînant un arrêt de prolifération permanent est également observé in vivo dans différents systèmes, mais n’a pas encore été étudié dans le contexte de la niche hématopoïétique. Les travaux présentés dans cette thèse ont pour objectif de déterminer si l’IR induit la sénescence des cellules stromales de la moelle osseuse et si une telle induction altère les fonctions hématopoïétiques. Nos résultats ont permis de démontrer pour la première fois qu’une IR corporelle totale induit effectivement la sénescence des cellules stromales de la moelle osseuse. En outre, cette altération du microenvironnement affecte la lymphopoïèse B de façon Ink4a/Arf-dépendante (1er article). De plus, les modifications systémiques qui résultent de l’IR compromettent l’homéostasie osseuse en augmentant la résorption de l’os, sans toutefois diminuer la formation de celui-ci (2e article). Ces données nous permettent de mieux comprendre les effets de la sénescence des cellules stromales de la moelle osseuse sur les fonctions hématopoïétiques. Par ailleurs, elles suggèrent que l’emploi de drogues et/ou de procédés n’induisant pas la sénescence des cellules stromales de l’os offrirait un meilleur pronostic à long terme pour les patients. / Ionizing radiation (IR) is used in the treatment of several cancers and hematological disorders, especially in conditioning regimens for bone marrow transplantation. Reduced doses of IR seem to favor the success of engraftment. This observation supports the growing evidences suggesting the importance of the microenvironment integrity for the success of bone marrow transplantation and hematopoiesis maintenance. IR induces senescence of bone marrow stromal cells in vitro. This defense mechanism which leads to a permanent cell growth arrest is also observed in different organs in vivo but has not yet been studied in the hematopoietic niche. The objectives of this doctoral thesis are to determine whether IR induces senescence of bone marrow stromal cells and whether such induction alters hematopoietic functions. Our results have demonstrated for the first time that total body IR actually induces the senescence of bone marrow stromal cells. Furthermore, this alteration of the microenvironment affects B lymphopoiesis in an Ink4a/Arf-dependent manner (paper #1). In addition, the systemic changes associated with IR compromise bone homeostasis by increasing bone resorption without reducing bone formation (paper #2). All together, these data enhance our knowledge related to the effects of IR-induced senescent bone marrow stromal cells on hematopoietic function. Moreover, our results suggest that using drugs and/or procedures inducing no senescent bone marrow stromal cells would provide a better long-term prognosis for patients.
274

Intranuclear Trafficking of RUNX/AML/CBFA/PEBP2 Transcription Factors in Living Cells: A Dissertation

Harrington, Kimberly Stacy 28 March 2003 (has links)
The family of runt related transcription factors (RUNX/Cbfa/AML/PEBP2) are essential for cellular differentiation and fetal development. RUNX factors are distributed throughout the nucleus in punctate foci that are associated with the nuclear matrix/scaffold and generally correspond with sites of active transcription. Truncations of RUNX proteins that eliminate the C-terminus including a 31-amino acid segment designated the nuclear matrix targeting signal (NMTS) lose nuclear matrix association and result in lethal hematopoietic (RUNX1) and skeletal (RUNX2) phenotypes in mice. These findings suggest that the targeting of RUNX factors to subnuclear foci may mediate the formation of multimeric regulatory complexes and contribute to transcriptional control. In this study, we hypothesized that RUNX transcription factors may dynamically move through the nucleus and associate with subnuclear domains in a C-terminal dependent mechanism to regulate transcription. Therefore, we investigated the subnuclear distribution and mobility of RUNX transcription factors in living cells using enhanced green fluorescent protein (EGFP) fused to RUNX proteins. The RUNX C-terminus was demonstrated to be necessary for the dynamic association of RUNX with stable subnuclear domains. Time-lapse fluorescence microscopy showed that RUNX1 and RUNX2 localize to punctate foci that remain stationary in the nuclear space in living cells. By measuring fluorescence recovery after photobleaching, both RUNX1 and RUNX2 were found to dynamically and rapidly associate with these subnuclear foci with a half-time of recovery in the ten-second time scale. A large immobile fraction of RUNX1 and RUNX2 proteins was observed in the photobleaching experiments, which suggests that this fraction of RUNX1 and RUNX2 proteins are immobilized through the C-terminal domain by interacting with the nuclear architecture. Truncation of the C-terminus of RUNX2, which removes the NMTS as well as several co-regulatory protein interaction domains, increases the mobility of RUNX2 by at least an order of magnitude, resulting in a half-time of recovery equivalent to that of EGFP alone. Contributions of the NMTS sequence to the subnuclear distribution and mobility of RUNX2 were further assessed by creating point mutations in the NMTS of RUNX2 fused to EGFP. The results show that these point mutations decrease, but do not abolish, association with the nuclear matrix compared to wild-type EGFP-RUNX2. Three patterns of subnuclear distribution were similarly observed in living cells for both NMTS mutants and wild-type RUNX2. Furthermore, the NMTS mutations showed no measurable effect on the mobility of RUNX2. However, the mobility of RUNX proteins in each of the different subnuclear distributions observed in living cells were significantly different from each other. The punctate distribution appears to correlate with higher fluorescence intensity, suggesting that the protein concentration in the cell may have an effect on the formation or size of the foci. These findings suggest that the entire NMTS and/or the co-regulatory protein interaction domains may be necessary to immobilize RUNX2 proteins. Because RUNX factors contain a conserved intranuclear targeting signal, we examined whether RUNX1 and RUNX2 are targeted to common subnuclear domains. The results show that RUNX1 and RUNX2 colocalized in common subnuclear foci. Furthermore, RUNX subnuclear foci contain the co-regulatory protein CBFβ, which heterodimerizes with RUNX factors, and nascent transcripts as shown by BrUTP incorporation. These results suggest that RUNX subnuclear foci may represent sites of transcription containing multi-subunit transcription factor complexes. RUNX2 transcription factors induce expression of the osteocalcin promoter during osteoblast differentiation and to study both RUNX2 and osteocalcin function, it would be helpful to have transgenic mice in which OC expression could be easily evaluated. Therefore, to assess the in vivo regulation of osteocalcin by RUNX protein, we generated transgenic mice expressing EGFP controlled by the osteocalcin promoter. Our results show that EGFP is expressed from the OC promoter in a cultured osteosarcoma cell line, but not in a kidney cell line, and is induced by vitamin D3. Furthermore, the OC-EGFP transgenic mice specifically express EGFP in osteoblasts and osteocytes in bone tissues. Moreover, EGFP is expressed in mineralized bone nodules of differentiated bone marrow derived from transgenic mice. Thus, these mice produce a good model for studying the in vivo effects of RUNX-mediated osteocalcin regulation and for developing potential drug therapies for bone diseases. Taken together, our results in living cells support the conclusion that RUNX transcription factors dynamically associate with stationary subnuclear foci in a C-terminal dependent mechanism to regulate gene expression. Moreover, RUNX subnuclear foci represent transcription sites containing nascent transcripts and co-regulatory interacting proteins. These conclusions provide a mechanism for how RUNX transcription factors may associate with subnuclear foci to regulate gene expression. Furthermore, the OC-EGFP transgenic mice now provide a useful tool for studying the in vivo function and regulation of osteocalcin by RUNX proteins during osteoblast differentiation and possibly for developing therapeutic drugs for treatment of bone diseases in the future.
275

Prostaglandin E₂ promotes recovery of hematopoietic stem and progenitor cells after radiation exposure

Stilger, Kayla N. 11 July 2014 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / The hematopoietic system is highly proliferative, making hematopoietic stem and progenitor cells (HSPC) sensitive to radiation damage. Total body irradiation and chemotherapy, as well as the risk of radiation accident, create a need for countermeasures that promote recovery of hematopoiesis. Substantive damage to the bone marrow from radiation exposure results in the hematopoietic syndrome of the acute radiation syndrome (HS-ARS), which includes life-threatening neutropenia, lymphocytopenia, thrombocytopenia, and possible death due to infection and/or hemorrhage. Given adequate time to recover, expand, and appropriately differentiate, bone marrow HSPC may overcome HS-ARS and restore homeostasis of the hematopoietic system. Prostaglandin E2 (PGE2) is known to have pleiotropic effects on hematopoiesis, inhibiting apoptosis and promoting self-renewal of hematopoietic stem cells (HSC), while inhibiting hematopoietic progenitor cell (HPC) proliferation. We assessed the radiomitigation potential of modulating PGE2 signaling in a mouse model of HS-ARS. Treatment with the PGE2 analog 16,16 dimethyl PGE2 (dmPGE2) at 24 hours post-irradiation resulted in increased survival of irradiated mice compared to vehicle control, with greater recovery in HPC number and colony-forming potential measured at 30 days post-irradiation. In a sublethal mouse model of irradiation, dmPGE2-treatment at 24 hours post-irradiation is associated with enhanced recovery of HSPC populations compared to vehicle-treated mice. Furthermore, dmPGE2-treatment may also act to promote recovery of the HSC niche through enhancement of osteoblast-supporting megakaryocyte (MK) migration to the endosteal surface of bone. A 2-fold increase in MKs within 40 um of the endosteum of cortical bone was seen at 48 hours post-irradiation in mice treated with dmPGE2 compared to mice treated with vehicle control. Treatment with the non-steroidal anti-inflammatory drug (NSAID) meloxicam abrogated this effect, suggesting an important role for PGE2 signaling in MK migration. In vitro assays support this data, showing that treatment with dmPGE2 increases MK expression of the chemokine receptor CXCR4 and enhances migration to its ligand SDF-1, which is produced by osteoblasts. Our results demonstrate the ability of dmPGE2 to act as an effective radiomitigative agent, promoting recovery of HSPC number and enhancing migration of MKs to the endosteum where they play a valuable role in niche restoration.
276

The essential role of Stat3 in bone homeostasis and mechanotransduction

Zhou, Hongkang January 2014 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Signal Transducer and Activator of Transcription 3 (Stat3) is a transcription factor expressed in bone and joint cells that include osteoblasts, osteocytes, osteoclasts, and chondrocytes. Stat3 is activated by a variety of cytokines and growth factors, including IL-6/gp130 family cytokines. These cytokines not only regulate the differentiation of osteoblasts and osteoclasts, but also regulate proliferation of chondrocytes through Stat3 activation. In 2007, mutations of Stat3 have been confirmed to cause a rare human immunodeficiency disease – Job syndrome which presents skeletal abnormalities like: reduced bone density (osteopenia), scoliosis, hyperextensibility of joints, and recurrent pathological bone fractures. Changes in the Stat3 gene alter the structure and function of the Stat3 proteins, impairing its ability to control the activity of other genes. However, little is known about the effects of Stat3 mutations on bone cells and tissues. To investigate the in vivo physiological role of Stat3 in bone homeostasis, osteoblast/osteocyte-specific Stat3 knockout (KO) mice were generated via the Cre-LoxP recombination system. The osteoblast/osteocyte-specific Stat3 KO mice showed bone abnormalities and an osteoporotic phenotype because of a reduced bone formation rate. Furthermore, inactivation of Stat3 decreased load-driven bone formation, and the disruption of Stat3 in osteoblasts suppressed load-driven mitochondrial activity, which led to an elevated level of reactive oxygen species (ROS) in cultured primary osteoblasts. Stat3 has been found to be responsive to mechanical stimulation, and might play an important role in mechanical signal transduction in osteocytes. To investigate the role Stat3 plays in mechanical signaling transduction, osteocyte-specific Stat3 knockout (KO) mice were created. Inactivation of Stat3 in osteocytes presented a significantly reduced load-driven bone formation. Decreased osteoblast activity indicated by reduced osteoid surface was also found in osteocyte-specific Stat3 KO mice. Moreover, sclerostin (SOST) protein which is a critical osteocyte-specific inhibitor of bone formation, its encoded gene SOST expression has been found to be enhanced in osteocyte-specific Stat3 KO mice. Thus, these results clearly demonstrated that Stat3 plays an important role in bone homeostasis and mechanotransduction, and Stat3 is not only involved in bone-formation-important genes regulation in the nucleus but also in mediation of ROS and oxidative stress in mitochondria.

Page generated in 0.0545 seconds