44 research outputs found

    Production of carotenoids by Rhodotorula mucilaginosa using sugarcane juice (Saccharum officinarum) in the fermentation. / Produção de carotenóides pela Rhodotorula mucilaginosa utilizando sumo de cana (Saccharum officinarum) na fermentação.

    Get PDF
    This study aims to investigate the production of carotenoids from pigmented yeasts isolated from Bahia’s semi-arid region, using sugarcane juice as an alternative fermentation medium.  The 23 full factorial statistical design was used to test the following independent variables: concentration of sugarcane juice (%) and yeast extract (g L-1). The Doehlert design was used to test the variables of initial pH and stirring speed. The maximum production obtained showed a concentration of 40% sugarcane juice, 6.0 g L-1 yeast extract, initial pH of 6.8 and stirring speed of 176 rpm, which enabled a carotenoid production of 1300 ?g L-1. As for the cell growth, the maximum was obtained at the concentration of 43% sugarcane juice, 9.2 g L-1 yeast extract, with an initial pH of 6.8 and stirring speed of 176 rpm, resulting in production of 14.7 g L-1. The HPLC analysis showed the presence of ?-carotene among the compounds produced by the yeast. The results showed the use sugarcane supplemented with yeast extract and by controlling environmental conditions such as initial pH and stirring speed, it is possible to promote an increase in the bioproduction of carotenoids and biomass of R. mucilaginosa

    Caspase-8 mediates inflammation and disease in rodent malaria

    Get PDF
    Earlier studies indicate that either the canonical or non-canonical pathways of inflammasome activation have a limited role on malaria pathogenesis. Here, we report that caspase-8 is a central mediator of systemic inflammation, septic shock in the Plasmodium chabaudi-infected mice and the P. berghei-induced experimental cerebral malaria (ECM). Importantly, our results indicate that the combined deficiencies of caspases-8/1/11 or caspase-8/gasdermin-D (GSDM-D) renders mice impaired to produce both TNFalpha and IL-1beta and highly resistant to lethality in these models, disclosing a complementary, but independent role of caspase-8 and caspases-1/11/GSDM-D in the pathogenesis of malaria. Further, we find that monocytes from malaria patients express active caspases-1, -4 and -8 suggesting that these inflammatory caspases may also play a role in the pathogenesis of human disease

    GM-CSF Priming Drives Bone Marrow-Derived Macrophages to a Pro-Inflammatory Pattern and Downmodulates PGE(2) in Response to TLR2 Ligands

    Get PDF
    In response to pathogen recognition by Toll-like receptors (TLRs) on their cell surface, macrophages release lipid mediators and cytokines that are widely distributed throughout the body and play essential roles in host responses. Granulocyte macrophage colony-stimulating factor (GM-CSF) is important for the immune response during infections to improve the clearance of microorganisms. In this study, we examined the release of mediators in response to TLR2 ligands by bone marrow-derived macrophages (BMDMs) primed with GM-CSF. We demonstrated that when stimulated with TLR2 ligands, non-primed BMDMs preferentially produced PGE(2) in greater amounts than LTB4. However, GM-CSF priming shifted the release of lipid mediators by BMDMs, resulting in a significant decrease of PGE(2) production in response to the same stimuli. The decrease of PGE(2) production from primed BMDMs was accompanied by a decrease in PGE-synthase mRNA expression and an increase in TNF-alpha and nitric oxide (NO) production. Moreover, some GM-CSF effects were potentiated by the addition of IFN-gamma. Using a variety of TLR2 ligands, we established that PGE(2) release by GM-CSF-primed BMDMs was dependent on TLR2 co-receptors (TLR1, TLR6), CD14, MyD88 and the nuclear translocation of NF kappa B but was not dependent on peroxisome proliferator-activated receptor-gamma (PPAR-gamma) activation. Indeed, GM-CSF priming enhanced TLR2, TLR4 and MyD88 mRNA expression and phospho-I kappa B alpha formation. These findings demonstrate that GM-CSF drives BMDMs to present a profile relevant to the host during infections.Fundacao de Amparo a Pesquisa do Estado de Sao Paulo (FAPESP)FAPESP - Fundacao de Amparo a Pesquisa do Estado de Sao PauloConselho Nacional de Pesquisa (CNPq)CNPq - Conselho Nacional de Pesquis

    In vitro effects of European and Latin-American medicinal plants in CYP3A4 gene expression, glutathione levels, and P-glycoprotein activity

    Get PDF
    Many medicinal plants species from European -such as Artemisia absinthium, Equisetum arvense, Lamium album, Malva sylvestris, Morus nigra, Passiflora incarnata, Frangula purshiana, and Salix alba- as well as Latin American traditions -such as Libidibia ferrea, Bidens pilosa, Casearia sylvestris, Costus spicatus, Monteverdia ilicifolia, Persea americana, Schinus terebinthifolia, Solidago chilensis, Syzygium cumini, Handroanthus impetiginosus, and Vernonanthura phosphorica- are shortlisted by the Brazilian National Health System for future clinical use. However, they lack many data on their action upon some key ADME targets. In this study, we assess non-toxic concentrations (up to100 μg/ml) of their infusions for in vitro ability to modulate CYP3A4 mRNA gene expression and intracellular glutathione levels in HepG2 cells, as well as P-glycoprotein (P-gp) activity in vincristine-resistant Caco-2 cells (Caco-2 VCR). We further investigated the activation of human pregnane X receptor (hPXR) in transiently co-transfected HeLa cells and the inhibition of Gamma-glutamyl transferase (GGT) in HepG2 cells. Our results demonstrate L. ferrea, C. sylvestris, M. ilicifolia, P. americana, S. terebinthifolia, S. cumini, V. phosphorica, E. arvense, P. incarnata, F. purshiana, and S. alba can significantly increase CYP3A4 mRNA gene expression in HepG2 cells. Only F. purshiana shown to do so likely via hPXR activation. P-gp activity was affected by L. ferrea, F. purshiana, S. terebinthifolia, and S. cumini. Total intracellular glutathione levels were significantly depleted by exposure to all extracts except S. alba and S. cumini This was accompanied by a lower GGT activity in the case of C. spicatus, P. americana, S. alba, and S. terebinthifolia, whilst L. ferrea, P. incarnata and F. purshiana increased it. Surprisingly, S. cumini aqueous extract drastically decreased GGT activity (−48%, p < 0.01). In conclusion, this preclinical study shows that the administration of some of these herbal medicines causes in vitro disturbances to key drug metabolism mechanisms. We recommend active pharmacovigilance for Libidibia ferrea (Mart.) L. P. Queiroz, Frangula purshiana Cooper, Schinus terebinthifolia Raddi, and Salix alba L. which were able to alter all targets in our preclinical study

    Pervasive gaps in Amazonian ecological research

    Get PDF
    Biodiversity loss is one of the main challenges of our time,1,2 and attempts to address it require a clear un derstanding of how ecological communities respond to environmental change across time and space.3,4 While the increasing availability of global databases on ecological communities has advanced our knowledge of biodiversity sensitivity to environmental changes,5–7 vast areas of the tropics remain understudied.8–11 In the American tropics, Amazonia stands out as the world’s most diverse rainforest and the primary source of Neotropical biodiversity,12 but it remains among the least known forests in America and is often underrepre sented in biodiversity databases.13–15 To worsen this situation, human-induced modifications16,17 may elim inate pieces of the Amazon’s biodiversity puzzle before we can use them to understand how ecological com munities are responding. To increase generalization and applicability of biodiversity knowledge,18,19 it is thus crucial to reduce biases in ecological research, particularly in regions projected to face the most pronounced environmental changes. We integrate ecological community metadata of 7,694 sampling sites for multiple or ganism groups in a machine learning model framework to map the research probability across the Brazilian Amazonia, while identifying the region’s vulnerability to environmental change. 15%–18% of the most ne glected areas in ecological research are expected to experience severe climate or land use changes by 2050. This means that unless we take immediate action, we will not be able to establish their current status, much less monitor how it is changing and what is being lostinfo:eu-repo/semantics/publishedVersio

    Pervasive gaps in Amazonian ecological research

    Get PDF

    Pervasive gaps in Amazonian ecological research

    Get PDF
    Biodiversity loss is one of the main challenges of our time,1,2 and attempts to address it require a clear understanding of how ecological communities respond to environmental change across time and space.3,4 While the increasing availability of global databases on ecological communities has advanced our knowledge of biodiversity sensitivity to environmental changes,5,6,7 vast areas of the tropics remain understudied.8,9,10,11 In the American tropics, Amazonia stands out as the world's most diverse rainforest and the primary source of Neotropical biodiversity,12 but it remains among the least known forests in America and is often underrepresented in biodiversity databases.13,14,15 To worsen this situation, human-induced modifications16,17 may eliminate pieces of the Amazon's biodiversity puzzle before we can use them to understand how ecological communities are responding. To increase generalization and applicability of biodiversity knowledge,18,19 it is thus crucial to reduce biases in ecological research, particularly in regions projected to face the most pronounced environmental changes. We integrate ecological community metadata of 7,694 sampling sites for multiple organism groups in a machine learning model framework to map the research probability across the Brazilian Amazonia, while identifying the region's vulnerability to environmental change. 15%–18% of the most neglected areas in ecological research are expected to experience severe climate or land use changes by 2050. This means that unless we take immediate action, we will not be able to establish their current status, much less monitor how it is changing and what is being lost

    Analysis of the antiangiogenic and antiproliferative effects of halofuginone on acute promyelocytic leukemia

    No full text
    Angiogênese é o termo utilizado para descrever o crescimento de novos vasos sanguíneos a partir dos já existentes. Vários estudos demonstraram a densidade microvascular (DMV) como um fator prognóstico nas leucemias, em particular, leucemia promielocítica aguda (LPA). Este subtipo de leucemia corresponde a cerca de 20 a 25% das leucemias mielóides agudas nos países latino-americanos e apresenta características clínicas, morfológicas e biológicas peculiares. A halofuginona (HF), originalmente descrita como um agente antifúngico apresenta capacidade de inibir o crescimento tumoral e formação de vasos em modelos animais de tumores sólidos. Estudos realizados por nosso grupo demonstraram que a HF inibiu a secreção do VEGF e a proliferação de linhagens celulares de LPA. Desta forma, o presente trabalho teve por objetivo determinar o potencial antiproliferativo e antiangiogênico da HF em um modelo experimental in vivo da LPA e avaliar os mecanismos subjacentes a sua ação. Primeiramente, a análise do ciclo celular em células NB4 tratadas com HF apresentou significativa diminuição da proliferação celular (2,093 ± 0,304 vs. 41,21 ± 3,25), juntamente com um aumento significativo da apoptose (12,53 ± 1,53 vs. 21,95 ± 0,79; p=0,0007). Por meio da técnica de Real Time Array foi possível identificar dois grupos de genes associados a apoptose celular diferencialmente expressos em células tratadas com HF: TNF, TNFRSF9, TNFTSF10B, CD40, FAS, CASP10, CASP8 e CASP3, sugerindo que a HF induz a via extrínseca de apoptose. A análise in vivo da HF foi realizada em camundongos NOD/SCID previamente irradiados e transplantados com células leucêmicas PML-RAR murinas. Camundongos tratados com HF, por 21 dias após o transplante não apresentaram remissão molecular, determinada pela amplificação do gene PML-RARA por PCR, porém foi observada menor infiltração leucêmica em relação aos camundongos não tratados (Leucócitos: 4,2 ± 3,89 vs. 20,6 ± 21,9; p<0.0001); Hemoglobina: 12,0 ± 1,40 vs. 9,6 ± 1,67; p<0.0001; e Plaquetas: 932,0 ± 122,5 vs. 552,0 ± 83,2; p<0.001 respectivamente) e um menor peso relativo do baço (0,006 vs. 0,012, p=0,0415). Ademais, a contagem diferencial e imunofenotipagem da medula óssea evidenciaram menor porcentagem de células mielóides imaturas (16,88 ± 6,27 vs. 44,06 ± 27,06). A HF também foi capaz de inibir a fosforilação de SMAD2 e consequentemente bloquear a via do TGF- em células NB4. No entanto, animais leucêmicos apresentaram menor nível sérico de TGF- em relação aos saudáveis e tratados (475,58 vs. 1.378,45/1.146,82 pg/mL; p<0,0001), sugerindo que o blasto leucêmico produz esta citocina e a diminuição de células leucêmicas resultou em diminuição dos níveis séricos de TGF-. A HF não aumentou a sobrevida dos animais leucêmicos e a elevação das enzimas hepáticas sugeriu que o tratamento foi hepatotóxico. Por fim, com relação à angiogênese, a análise da expressão gênica mostrou que o tratamento com HF inibiu a expressão de VEGF e EGF e o estudo por imunohistoquímica de seções da medula óssea evidenciou menor expressão VEGF (30 vs. 80%, p=0,0227), porém não houve diminuição da DMV. O conjunto desses resultados mostrou que a angiogênese é um importante alvo terapêutico na LPA, e que apesar da toxicidade, a HF apresenta potencial antileucêmico, tanto por conta de seus efeitos antiproliferativos e próapoptóticos, quanto por sua capacidade de inibir a produção de fatores próangiogênicos.Angiogenesis is the term used to describe the growth of new blood vessels from the existing ones. Several studies have demonstrated the microvascular density (MVD) as a prognostic factor in leukemia, particularly acute promyelocytic leukemia (APL). This subtype of leukemia corresponds to 20-25% of acute myeloid leukemia in Latin America and presents clinical, morphological and biological peculiar characteristics. The halofuginone (HF) originally described as an antifungal agent has ability to inhibit tumor growth and vessel formation in animal models of solid tumors. Our group demonstrated that HF inhibits the VEGF secretion and cell proliferation in APL cell lineages. Thus, this study aimed to determine the antiproliferative and antiangiogenic potential of HF in an experimental model of APL in vivo and evaluate the mechanisms underlying its action. First, the cell cycle analysis in NB4 cells treated with HF showed a significant decrease in cell proliferation (2.093 ± 0.304 vs. 41.21 ± 3.25), along with a significant increase in apoptosis (12.53 ± 1.53 vs . 21.95 ± 0.79, p = 0.0007). Through Real Time Array it was possible to identify two groups of apoptosis associated genes differentially expressed in HF treated cells: TNF, TNFRSF9, TNFTSF10B, CD40, FAS, CASP10, CASP8 and CASP3, suggesting that HF induces apoptosis by extrinsic pathway. In vivo analysis of HF was performed in irradiated NOD/SCID mice transplanted with murine PML-RAR leukemic cells. Mice treated with HF for 21 days after transplantation showed no molecular remission as determined by amplification of PML-RARA gene by PCR, however minor leukemic infiltration was observed compared to untreated mice (leukocytes: 4.2 ± 3.89 vs . 20.6 ± 21.9, p <0.0001), hemoglobin: 12.0 ± 1.40 vs. 9.6 ± 1.67, p <0.0001, and Platelets: 932.0 ± 122.5 vs. 552.0 ± 83.2, p <0.001 respectively) and a lower relative weight of spleen (0.006 vs. 0.012, p = 0.0415). Furthermore, the differential count and immunophenotyping of bone marrow showed a lower percentage of immature myeloid cells (16.88 ± 6.27 vs. 44.06 ± 27.06). The HF was also able to inhibit the SMAD2 phosphorylation and consequently block the TGF- pathway in NB4 cells. However, leukemic animals presented lower serum TGF- compared to the healthy and treated (475.58 vs. 1378.45/1146.82 pg / mL, p <0.0001), suggesting that the leukemic blasts produces this cytokines and the decrease in leukemic cells resulted in decreased serum levels of TGF-. The HF did not increase the survival of leukemic animals and elevated liver enzymes suggested that the treatment was hepatotoxic. Finally, regarding angiogenesis, gene expression analysis showed that the HF treatment inhibited the expression of VEGF and EGF and the study by immunohistochemistry of sections of bone marrow showed less VEGF expression (30 vs. 80%, p = 0 0227), but there was no decrease in the MVD. Taken together, these results showed that angiogenesis is an important therapeutic target in APL, and despite the toxicity, HF has antileukemic potential, for both the antiproliferative and proapoptotic effects, and also for its ability to inhibit the production of proangiogenic factors

    Analysis of the antiangiogenic and antiproliferative effects of halofuginone on acute promyelocytic leukemia

    No full text
    Angiogênese é o termo utilizado para descrever o crescimento de novos vasos sanguíneos a partir dos já existentes. Vários estudos demonstraram a densidade microvascular (DMV) como um fator prognóstico nas leucemias, em particular, leucemia promielocítica aguda (LPA). Este subtipo de leucemia corresponde a cerca de 20 a 25% das leucemias mielóides agudas nos países latino-americanos e apresenta características clínicas, morfológicas e biológicas peculiares. A halofuginona (HF), originalmente descrita como um agente antifúngico apresenta capacidade de inibir o crescimento tumoral e formação de vasos em modelos animais de tumores sólidos. Estudos realizados por nosso grupo demonstraram que a HF inibiu a secreção do VEGF e a proliferação de linhagens celulares de LPA. Desta forma, o presente trabalho teve por objetivo determinar o potencial antiproliferativo e antiangiogênico da HF em um modelo experimental in vivo da LPA e avaliar os mecanismos subjacentes a sua ação. Primeiramente, a análise do ciclo celular em células NB4 tratadas com HF apresentou significativa diminuição da proliferação celular (2,093 ± 0,304 vs. 41,21 ± 3,25), juntamente com um aumento significativo da apoptose (12,53 ± 1,53 vs. 21,95 ± 0,79; p=0,0007). Por meio da técnica de Real Time Array foi possível identificar dois grupos de genes associados a apoptose celular diferencialmente expressos em células tratadas com HF: TNF, TNFRSF9, TNFTSF10B, CD40, FAS, CASP10, CASP8 e CASP3, sugerindo que a HF induz a via extrínseca de apoptose. A análise in vivo da HF foi realizada em camundongos NOD/SCID previamente irradiados e transplantados com células leucêmicas PML-RAR murinas. Camundongos tratados com HF, por 21 dias após o transplante não apresentaram remissão molecular, determinada pela amplificação do gene PML-RARA por PCR, porém foi observada menor infiltração leucêmica em relação aos camundongos não tratados (Leucócitos: 4,2 ± 3,89 vs. 20,6 ± 21,9; p<0.0001); Hemoglobina: 12,0 ± 1,40 vs. 9,6 ± 1,67; p<0.0001; e Plaquetas: 932,0 ± 122,5 vs. 552,0 ± 83,2; p<0.001 respectivamente) e um menor peso relativo do baço (0,006 vs. 0,012, p=0,0415). Ademais, a contagem diferencial e imunofenotipagem da medula óssea evidenciaram menor porcentagem de células mielóides imaturas (16,88 ± 6,27 vs. 44,06 ± 27,06). A HF também foi capaz de inibir a fosforilação de SMAD2 e consequentemente bloquear a via do TGF- em células NB4. No entanto, animais leucêmicos apresentaram menor nível sérico de TGF- em relação aos saudáveis e tratados (475,58 vs. 1.378,45/1.146,82 pg/mL; p<0,0001), sugerindo que o blasto leucêmico produz esta citocina e a diminuição de células leucêmicas resultou em diminuição dos níveis séricos de TGF-. A HF não aumentou a sobrevida dos animais leucêmicos e a elevação das enzimas hepáticas sugeriu que o tratamento foi hepatotóxico. Por fim, com relação à angiogênese, a análise da expressão gênica mostrou que o tratamento com HF inibiu a expressão de VEGF e EGF e o estudo por imunohistoquímica de seções da medula óssea evidenciou menor expressão VEGF (30 vs. 80%, p=0,0227), porém não houve diminuição da DMV. O conjunto desses resultados mostrou que a angiogênese é um importante alvo terapêutico na LPA, e que apesar da toxicidade, a HF apresenta potencial antileucêmico, tanto por conta de seus efeitos antiproliferativos e próapoptóticos, quanto por sua capacidade de inibir a produção de fatores próangiogênicos.Angiogenesis is the term used to describe the growth of new blood vessels from the existing ones. Several studies have demonstrated the microvascular density (MVD) as a prognostic factor in leukemia, particularly acute promyelocytic leukemia (APL). This subtype of leukemia corresponds to 20-25% of acute myeloid leukemia in Latin America and presents clinical, morphological and biological peculiar characteristics. The halofuginone (HF) originally described as an antifungal agent has ability to inhibit tumor growth and vessel formation in animal models of solid tumors. Our group demonstrated that HF inhibits the VEGF secretion and cell proliferation in APL cell lineages. Thus, this study aimed to determine the antiproliferative and antiangiogenic potential of HF in an experimental model of APL in vivo and evaluate the mechanisms underlying its action. First, the cell cycle analysis in NB4 cells treated with HF showed a significant decrease in cell proliferation (2.093 ± 0.304 vs. 41.21 ± 3.25), along with a significant increase in apoptosis (12.53 ± 1.53 vs . 21.95 ± 0.79, p = 0.0007). Through Real Time Array it was possible to identify two groups of apoptosis associated genes differentially expressed in HF treated cells: TNF, TNFRSF9, TNFTSF10B, CD40, FAS, CASP10, CASP8 and CASP3, suggesting that HF induces apoptosis by extrinsic pathway. In vivo analysis of HF was performed in irradiated NOD/SCID mice transplanted with murine PML-RAR leukemic cells. Mice treated with HF for 21 days after transplantation showed no molecular remission as determined by amplification of PML-RARA gene by PCR, however minor leukemic infiltration was observed compared to untreated mice (leukocytes: 4.2 ± 3.89 vs . 20.6 ± 21.9, p <0.0001), hemoglobin: 12.0 ± 1.40 vs. 9.6 ± 1.67, p <0.0001, and Platelets: 932.0 ± 122.5 vs. 552.0 ± 83.2, p <0.001 respectively) and a lower relative weight of spleen (0.006 vs. 0.012, p = 0.0415). Furthermore, the differential count and immunophenotyping of bone marrow showed a lower percentage of immature myeloid cells (16.88 ± 6.27 vs. 44.06 ± 27.06). The HF was also able to inhibit the SMAD2 phosphorylation and consequently block the TGF- pathway in NB4 cells. However, leukemic animals presented lower serum TGF- compared to the healthy and treated (475.58 vs. 1378.45/1146.82 pg / mL, p <0.0001), suggesting that the leukemic blasts produces this cytokines and the decrease in leukemic cells resulted in decreased serum levels of TGF-. The HF did not increase the survival of leukemic animals and elevated liver enzymes suggested that the treatment was hepatotoxic. Finally, regarding angiogenesis, gene expression analysis showed that the HF treatment inhibited the expression of VEGF and EGF and the study by immunohistochemistry of sections of bone marrow showed less VEGF expression (30 vs. 80%, p = 0 0227), but there was no decrease in the MVD. Taken together, these results showed that angiogenesis is an important therapeutic target in APL, and despite the toxicity, HF has antileukemic potential, for both the antiproliferative and proapoptotic effects, and also for its ability to inhibit the production of proangiogenic factors
    corecore