Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Sensors (Basel) ; 21(20)2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34695979

RESUMO

Applying georadar (GPR) technology for detecting underground utilities is an important element of the comprehensive assessment of the location and ground infrastructure status. These works are usually connected with the conducted investment processes or serialised inventory of underground fittings. The detection of infrastructure is also crucial in implementing the BIM technology, 3D cadastre, and planned network modernization works. GPR detection accuracy depends on the type of equipment used, the selected detection method, and external factors. The multitude of techniques used for localizing underground utilities and constantly growing accuracy demands resulting from the fact that it is often necessary to detect infrastructure under challenging conditions of dense urban development leads to the need to improve the existing technologies. The factor that motivated us to start research on assessing the precision and accuracy of ground penetrating radar detection was the need to ensure the appropriate accuracy, precision, and reliability of detecting underground utilities versus different methods and analyses. The results of the multi-variant GPR were subjected to statistical testing. Various analyses were also conducted, depending on the detection method and on the current soil parameters using a unique sensor probe. When planning detection routes, we took into account regular, established grids and tracked the trajectory of movement of the equipment using GNSS receivers (internal and external ones). Moreover, a specialist probe was used to evaluate the potential influence of the changing soil conditions on the obtained detection results. Our tests were conducted in a developed area for ten months. The results confirmed a strong correlation between the obtained accuracy and the measurement method used, while the correlation with the other factors discussed here was significantly weaker.


Assuntos
Radar , Solo , Reprodutibilidade dos Testes
2.
J Cell Mol Med ; 20(1): 134-46, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26515267

RESUMO

Evidence has accumulated that murine haematopoietic stem/progenitor cells (HSPCs) share several markers with the germline, a connection supported by recent reports that pituitary and gonadal sex hormones (SexHs) regulate development of murine HSPCs. It has also been reported that human HSPCs, like their murine counterparts, respond to certain SexHs (e.g. androgens). However, to better address the effects of SexHs, particularly pituitary SexHs, on human haematopoiesis, we tested for expression of receptors for pituitary SexHs, including follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL), as well as the receptors for gonadal SexHs, including progesterone, oestrogens, and androgen, on HSPCs purified from human umbilical cord blood (UCB) and peripheral blood (PB). We then tested the functionality of these receptors in ex vivo signal transduction studies and in vitro clonogenic assays. In parallel, we tested the effect of SexHs on human mesenchymal stromal cells (MSCs). Finally, based on our observation that at least some of the UCB-derived, CD45(-) very small embryonic-like stem cells (VSELs) become specified into CD45(+) HSPCs, we also evaluated the expression of pituitary and gonadal SexH receptors on these cells. We report for the first time that human HSPCs and VSELs, like their murine counterparts, express pituitary and gonadal SexH receptors at the mRNA and protein levels. Most importantly, SexH if added to suboptimal doses of haematopoietic cytokines and growth factors enhance clonogenic growth of human HSPCs as well as directly stimulate proliferation of MSCs.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Esteroides/metabolismo , Adesão Celular , Proliferação de Células , Células Cultivadas , Sangue Fetal , Fibronectinas/metabolismo , Hormônios Esteroides Gonadais/fisiologia , Humanos
3.
J Cell Mol Med ; 19(9): 2193-201, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26033571

RESUMO

The glycolipid glycosylphosphatidylinositol anchor (GPI-A) plays an important role in lipid raft formation, which is required for proper expression on the cell surface of two inhibitors of the complement cascade, CD55 and CD59. The absence of these markers from the surface of blood cells, including erythrocytes, makes the cells susceptible to complement lysis, as seen in patients suffering from paroxysmal nocturnal haemoglobinuria (PNH). However, the explanation for why PNH-affected hematopoietic stem/progenitor cells (HSPCs) expand over time in BM is still unclear. Here, we propose an explanation for this phenomenon and provide evidence that a defect in lipid raft formation in HSPCs leads to defective CXCR4- and VLA-4-mediated retention of these cells in BM. In support of this possibility, BM-isolated CD34(+) cells from PNH patients show a defect in the incorporation of CXCR4 and VLA-4 into membrane lipid rafts, respond weakly to SDF-1 stimulation, and show defective adhesion to fibronectin. Similar data were obtained with the GPI-A(-) Jurkat cell line. Moreover, we also report that chimeric mice transplanted with CD55(-/-)  CD59(-/-) BM cells but with proper GPI-A expression do not expand over time in transplanted hosts. On the basis of these findings, we propose that a defect in lipid raft formation in PNH-mutated HSPCs makes these cells more mobile, so that they expand and out-compete normal HSPCs from their BM niches over time.


Assuntos
Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/patologia , Microdomínios da Membrana/metabolismo , Animais , Antígenos CD/metabolismo , Toxinas Bacterianas/metabolismo , Medula Óssea/patologia , Adesão Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Fibronectinas/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Humanos , Integrina alfa4beta1/metabolismo , Células Jurkat , Microdomínios da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Stem Cells ; 32(9): 2502-15, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24806427

RESUMO

Activation of the complement cascade (CC) with myocardial infarction (MI) acutely initiates immune cell infiltration, membrane attack complex formation on injured myocytes, and exacerbates myocardial injury. Recent studies implicate the CC in mobilization of stem/progenitor cells and tissue regeneration. Its role in chronic MI is unknown. Here, we consider complement component C3, in the chronic response to MI. C3 knockout (KO) mice were studied after permanent coronary artery ligation. C3 deficiency exacerbated myocardial dysfunction 28 days after MI compared to WT with further impaired systolic function and LV dilation despite similar infarct size 24 hours post-MI. Morphometric analysis 28 days post-MI showed C3 KO mice had more scar tissue with less viable myocardium within the infarct zone which correlated with decreased c-kit(pos) cardiac stem/progenitor cells (CPSC), decreased proliferating Ki67(pos) CSPCs and decreased formation of new BrdU(pos) /α-sarcomeric actin(pos) myocytes, and increased apoptosis compared to WT. Decreased CSPCs and increased apoptosis were evident 7 days post-MI in C3 KO hearts. The inflammatory response with MI was attenuated in the C3 KO and was accompanied by attenuated hematopoietic, pluripotent, and cardiac stem/progenitor cell mobilization into the peripheral blood 72 hours post-MI. These results are the first to demonstrate that CC, through C3, contributes to myocardial preservation and regeneration in response to chronic MI. Responses in the C3 KO infer that C3 activation in response to MI expands the resident CSPC population, increases new myocyte formation, increases and preserves myocardium, inflammatory response, and bone marrow stem/progenitor cell mobilization to preserve myocardial function.


Assuntos
Complemento C3/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Animais , Proliferação de Células/fisiologia , Complemento C3/genética , Modelos Animais de Doenças , Ecocardiografia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Regeneração/fisiologia , Função Ventricular Esquerda/fisiologia
5.
J Cell Mol Med ; 18(9): 1797-806, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895014

RESUMO

The concept that bone marrow (BM)-derived cells may participate in neural regeneration remains controversial, and the identity of the specific cell type(s) involved remains unknown. We recently reported that the adult murine BM contains a highly mobile population of Sca-1(+) Lin(-) CD45(-) cells known as very small embryonic/epiblast-like stem cells (VSELs) that express several markers of pluripotency such as Oct-4. In the BM microenvironment, these cells are kept quiescent because of epigenetic modification of certain paternally imprinted genes. However, as reported, these cells can be mobilized in mice in an experimental model of stroke and express several genes involved in neurogenesis while circulating in peripheral blood (PB). In the current work, we employed a model of toxic brain damage, which is induced by administration of kainic acid, to see not only whether VSELs can be mobilized into PB in response to this neurotoxin, but, more importantly, whether they proliferate and expand in BM tissue. We report here for the first time that brain damage leads to activation and expansion of the BM pool of quiescent VSELs, which precedes their subsequent egress into PB. Harnessing these cells in neural tissue regeneration is currently one of the challenges in regenerative medicine.


Assuntos
Células da Medula Óssea/fisiologia , Encefalopatias/patologia , Proliferação de Células/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Ácido Caínico/toxicidade , Animais , Células da Medula Óssea/efeitos dos fármacos , Encefalopatias/induzido quimicamente , Movimento Celular , Células Cultivadas , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Citometria de Fluxo , Masculino , Camundongos Endogâmicos C57BL
6.
Stem Cells ; 31(3): 500-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23193025

RESUMO

Ceramide-1-phosphate (C1P) is a bioactive lipid that, in contrast to ceramide, is an antiapoptotic molecule released from cells that are damaged and "leaky." As reported recently, C1P promotes migration of hematopoietic cells. In this article, we tested the hypothesis that C1P released upon tissue damage may play an underappreciated role in chemoattraction of various types of stem cells and endothelial cells involved in tissue/organ regeneration. We show for the first time that C1P is upregulated in damaged tissues and chemoattracts bone marrow (BM)-derived multipotent stromal cells, endothelial progenitor cells, and very small embryonic-like stem cells. Furthermore, compared to other bioactive lipids, C1P more potently chemoattracted human umbilical vein endothelial cells and stimulated tube formation by these cells. C1P also promoted in vivo vascularization of Matrigel implants and stimulated secretion of stromal cell-derived factor-1 from BM-derived fibroblasts. Thus, our data demonstrate, for the first time, that C1P is a potent bioactive lipid released from damaged cells that potentially plays an important and novel role in recruitment of stem/progenitor cells to damaged organs and may promote their vascularization.


Assuntos
Movimento Celular/fisiologia , Ceramidas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Processos de Crescimento Celular/fisiologia , Ceramidas/biossíntese , Fatores Quimiotáticos/biossíntese , Fatores Quimiotáticos/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Medicina Regenerativa/métodos , Células-Tronco/metabolismo , Regulação para Cima
7.
Prostaglandins Other Lipid Mediat ; 104-105: 122-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22981511

RESUMO

Under steady-state conditions, hematopoietic stem/progenitor cells (HSPCs) egress from bone marrow (BM) and enter peripheral blood (PB) where they circulate at low levels. Their number in PB, however, increases significantly in several stress situations related to infection, organ/tissue damage, or strenuous exercise. Pharmacologically mediated enforced egress of HSPCs from the BM microenvironment into PB is called "mobilization", and this phenomenon has been exploited in hematological transplantology as a means to obtain HSPCs for hematopoietic reconstitution. In this review we will present the accumulated evidence that innate immunity, including the complement cascade and the granulocyte/monocyte lineage, and the PB plasma level of the bioactive lipid sphingosine-1-phosphate (S1P) together orchestrate this evolutionarily conserved mechanism that directs trafficking of HSPCs.


Assuntos
Anemia Falciforme/metabolismo , Proteínas do Sistema Complemento/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Anemia Falciforme/imunologia , Anemia Falciforme/patologia , Quimiotaxia/imunologia , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Granulócitos/imunologia , Granulócitos/metabolismo , Granulócitos/patologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Hemoglobinúria Paroxística/imunologia , Hemoglobinúria Paroxística/patologia , Humanos , Imunidade Inata , Lisofosfolipídeos/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Esfingosina/imunologia , Esfingosina/metabolismo
8.
Cell Transplant ; 25(2): 275-82, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26087465

RESUMO

We found that diurnal activation of the three evolutionarily ancient proteolytic cascades in peripheral blood (PB), namely, the complement, coagulation, and fibrinolytic cascades, late at night or in the early morning hours, precedes the diurnal release of hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into PB in wild-type mice. Moreover, activation of the distal part of the complement cascade (ComC), involving cleavage of the fifth component (C5), seems to play a crucial role in pharmacological mobilization of HSPCs. In order to shed more light on the role of diurnal rhythms in the egress of HSPCs, we studied diurnal changes in the number of circulating HSPCs in C5-deficient mice and did not observe diurnal changes in the number of these cells circulating in PB in C5(-/-) animals. Based on this finding, we conclude that activation of the distal part of the ComC, C5 cleavage, and release of C5a and desArgC5a are required in executing the diurnal release of HSPCs from BM into PB. Moreover, the fact that C5(-/-) mice still displayed normal activation of the coagulation and fibrinolytic cascades indicates that, of all the proteolytic cascades, the ComC is the dominant player regulating diurnal egress of HSPCs.


Assuntos
Medula Óssea/metabolismo , Movimento Celular/fisiologia , Ativação do Complemento/imunologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Complemento C5/deficiência , Complemento C5/genética , Complemento C5/metabolismo , Granulócitos/citologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Oncotarget ; 7(3): 3033-46, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26701888

RESUMO

We recently reported that normal hematopoietic stem cells express functional pituitary sex hormone (SexH) receptors. Here we report for the first time that pituitary-secreted gonadotrophins stimulate migration, adhesion, and proliferation of several human myeloid and lymphoid leukemia cell lines. Similar effects were observed after stimulation of human leukemic cell lines by gonadal SexHs. This effect seems to be direct, as the SexH receptors expressed by leukemic cells responded to stimulation by phosphorylation of MAPKp42/44 and AKTser473. Furthermore, in parallel studies we confirmed that human primary patient-derived AML and CML blasts also express several functional SexH receptors. These results shed more light on the potential role of SexHs in leukemogenesis and, in addition, provide further evidence suggesting a developmental link between hematopoiesis and the germline.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Gonadotropinas Hipofisárias/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mieloide Aguda/metabolismo , Hormônio Luteinizante/farmacologia , Receptores do Hormônio Hipofisário/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Células Jurkat , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Mieloides/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
10.
Oncotarget ; 6(22): 18819-28, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26299919

RESUMO

The α-chemokine stromal-derived factor 1 (SDF-1), which binds to the CXCR4 receptor, directs migration and homing of CXCR4+ hematopoietic stem/progenitor cells (HSPCs) to bone marrow (BM) stem cell niches. Nevertheless, it is also known that CXCR4-/- fetal liver-derived hematopoietic stem cells engraft into BM and that blockade of CXCR4 by its antagonist AMD3100 does not prevent engraftment of HSPCs. Because of this finding of SDF-1-CXCR4-independent BM homing, the unique role of SDF-1 in HSPC homing has recently been challenged. While SDF-1 is the only chemokine that chemoattracts HSPCs, other chemoattractants for these cells have recently been described, including the bioactive phosphosphingolipid sphingosine-1-phosphate (S1P). To address the potential role of S1P in homing of HSPCs to BM, we performed hematopoietic transplants into mice deficient in BM-expressed sphingosine kinase 1 (Sphk1-/-) using hematopoietic cells from normal control mice as well as cells from mice in which floxed CXCR4 (CXCR4fl/fl) was conditionally deleted. We observed the presence of a homing and engraftment defect in HSPCs of Sphk1-/- mice that was particularly profound after transplantation of CXCR4-/- BM cells. Thus, our results indicate that BM-microenvironment-expressed S1P plays a role in homing of HSPCs. They also support the concept that, in addition to the SDF-1-CXCR4 axis, other chemotactic axes are also involved in homing and engraftment of HSPCs.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Condicionamento Pré-Transplante/métodos , Animais , Quimiocina CXCL12/metabolismo , Feminino , Masculino , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Nicho de Células-Tronco/fisiologia
11.
Stem Cells Dev ; 24(8): 927-37, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25607657

RESUMO

Evidence has accumulated that hematopoietic stem progenitor cells (HSPCs) share several markers with the germline, a connection supported by reports that prolactin, androgens, and estrogens stimulate hematopoiesis. To address this issue more directly, we tested the expression of receptors for pituitary-derived hormones, such as follicle-stimulating hormone (FSH) and luteinizing hormone (LH), on purified murine bone marrow (BM) cells enriched for HSPCs and tested the functionality of these receptors in ex vivo signal transduction studies and in vitro clonogenic assays. We also tested whether administration of pituitary- and gonad-derived sex hormones (SexHs) increases incorporation of bromodeoxyuridine (BrdU) into HSPCs and expansion of hematopoietic clonogenic progenitors in mice and promotes recovery of blood counts in sublethally irradiated animals. We report for the first time that HSPCs express functional FSH and LH receptors and that both proliferate in vivo and in vitro in response to stimulation by pituitary SexHs. Furthermore, based on our observations that at least some of CD45(-) very small embryonic-like stem cells (VSELs) may become specified into CD45(+) HSPCs, we also evaluated the expression of pituitary and gonadal SexHs receptors on these cells and tested whether these quiescent cells may expand in vivo in response to SexHs administration. We found that VSELs express SexHs receptors and respond in vivo to SexHs stimulation, as evidenced by BrdU accumulation. Since at least some VSELs share several markers characteristic of migrating primordial germ cells and can be specified into HSPCs, this observation sheds new light on the BM stem cell hierarchy.


Assuntos
Células Germinativas/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Receptores Androgênicos/metabolismo , Receptores do FSH/metabolismo , Receptores do LH/metabolismo , Receptores da Prolactina/metabolismo , Animais , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/genética , Receptores do FSH/genética , Receptores do LH/genética , Receptores da Prolactina/genética
12.
Expert Opin Ther Targets ; 18(1): 95-107, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24188167

RESUMO

INTRODUCTION: A common feature of many types of cells is their responsiveness to chemotactic gradients of factors for which they express the corresponding receptors. The most studied chemoattractants so far are peptide-based growth factors and a family of cytokines endowed with strong chemotactic properties, called chemokines. However, additional evidence has accumulated that, in addition to these peptide-based chemoattractants, an important role in cell migration is played by bioactive lipids. AREAS COVERED: Solid evidence has accumulated that two bioactive phosphorylated sphingolipids that are derivatives of sphingolipid metabolism, namely sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), are potent chemoattractants for a variety of cells. In this review, we will discuss the effect of these two phosphorylated sphingolipids on the trafficking of normal and malignant cells, and, in particular, we will focus on their role in trafficking of normal hematopoietic stem/progenitor cells. Unlike other mediators, S1P under steady-state conditions maintain a steep gradient between interstitial fluid and peripheral blood and lymph across the endothelial barrier, which is important in the egress of cells from bone marrow. Both S1P and C1P may be upregulated in damaged tissues, which may result in reversal of this gradient. EXPERT OPINION: S1P and C1P are important regulators of the trafficking of normal and malignant cells, and modification of their biological effects will have important applications in optimizing stem cell mobilization and homing, tissue organ/regeneration, and preventing cancer metastasis.


Assuntos
Ceramidas/fisiologia , Lisofosfolipídeos/fisiologia , Neoplasias/metabolismo , Esfingosina/análogos & derivados , Células-Tronco/metabolismo , Transporte Biológico , Humanos , Neoplasias/patologia , Esfingosina/fisiologia , Células-Tronco/citologia
13.
Histol Histopathol ; 28(10): 1325-36, 2013 10.
Artigo em Inglês | MEDLINE | ID: mdl-23613169

RESUMO

It is well known that somatotrophic/insulin signaling affects lifespan in experimental animals. To study the effects of insulin-like growth factor-1 (IGF-1) plasma level on the morphology of major organs, we analyzed lung, heart, liver, kidney, bone marrow, and spleen isolated from 2-year-old growth hormone receptor knockout (GHR-KO) Laron dwarf mice (with low circulating plasma levels of IGF-1) and 6-month-old bovine growth hormone transgenic (bGHTg) mice (with high circulating plasma levels of IGF-1). The ages of the two mutant strains employed in our studies were selected based on their overall ~50% survival (Laron dwarf mice live up to ~4 years and bGHTg mice up to ~1 year). Morphological analysis of the organs of long-living 2-year-old Laron dwarf mice revealed a lower biological age for their organs compared with normal littermates, with more brown adipose tissue (BAT) surrounding the main body organs, lower levels of steatosis in liver, and a lower incidence of leukocyte infiltration in different organs. By contrast, the organs of 6-month-old, short-living bGHTg mice displayed several abnormalities in liver and kidney and a reduced content of BAT around vital organs.


Assuntos
Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/fisiologia , Tecido Adiposo Marrom/patologia , Animais , Bovinos , Densitometria , Fígado Gorduroso/patologia , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Hepatócitos/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/patologia , Leucócitos/citologia , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Plasma/metabolismo , Ratos , Baço/patologia , Fatores de Tempo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa