首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The mechanism responsible for the metabolism of vitamin A during hypervitaminosis is largely unknown. This study investigated hepatic 13C-retinol uptake in hypervitaminotic A rhesus monkeys. We hypothesized that individual retinyl esters would be enriched in 13C after a physiologic dose of 13C2-retinyl acetate, thus suggesting de novo in vivo hepatic retinol esterification. Male rhesus macaques (n = 16; 11.8 ± 2.9 y) each received 3.5 µmol 14, 15-13C2-retinyl acetate. Blood was drawn at baseline and 5 h and 2, 4, 7, 14, 21, and 28 d after administration. Liver biopsies were collected 7 d before and 2 d after dose administration (n = 4) and at 7, 14, and 28 d after dose administration (n = 4 per time point). 13C enrichments of retinol and retinyl esters HPLC-purified from liver samples were measured by using gas chromatography–combustion–isotope ratio mass spectrometry. 13C enrichment of total vitamin A and individual retinyl esters were significantly greater 2 d after dose administration compared with baseline levels. In contrast, the concentration of isolated retinyl esters did not always increase 2 d after treatment. Given that the liver biopsy site differed between monkeys, these data suggest that the accumulation of hepatic retinyl esters is a dynamic process that is better represented by combining analytical techniques. This sensitive methodology can be used to characterize vitamin A trafficking after physiologic doses of 13C-retinol. In this nonhuman primate model of hypervitaminosis A, hepatic retinyl esters continued to accumulate with high liver stores.Abbreviations: GCCIRMS, gas chromatography–combustion–isotope ratio mass spectrometry, IRMS, isotope ratio mass spectrometry, PS/LO, ratio of retinyl palmitate plus stearate to retinyl linoleate plus oleateVitamin A is critical for vision, reproduction, and cellular differentiation.18 All tissue vitamin A originates as dietary vitamin A, which is predominantly available as preformed vitamin A (that is, retinyl esters)3 and the provitamin A carotenoids. Retinyl esters are cleaved to retinol in the intestinal lumen, and unesterified retinol is absorbed into the enterocyte.25 Retinol is esterified within the intestinal mucosa23,31 and then packaged into chylomicrons. These particles are exocytosed and transported into the general circulation,4 where they are degraded to chylomicron remnants and taken up by the liver.5,9 Once in hepatic parenchymal cells, retinyl esters are rapidly hydrolyzed to retinol and, depending on the animals’ vitamin A status, retinol is secreted back into plasma bound to retinol-binding protein or transferred to stellate cells, reesterified, and stored.6 Thus, vitamin A in the liver occurs in 2 forms: as retinol and esterified to various fatty acids. Analysis of hepatic retinyl esters within 30 min of intravenous injection of labeled chylomicron retinyl ester in vitamin-A–sufficient rats recovered 80% to 90% of the dose. During vitamin A sufficiency, a majority of labeled chylomicron retinyl esters are taken up by the liver before subsequent hydrolysis.5Little is known about the storage and metabolism of vitamin A during hypervitaminosis A,32,49 despite the wide use of retinoids pharmaceutically.48 An early study involving hypervitaminotic A rats characterized increased concentrations of retinyl esters as percentages of total vitamin A in the plasma profile during excessive consumption of vitamin A.32 This study further identified that retinyl esters were transported in the serum by means of lipoproteins, thus mediating the vitamin''s nonspecific delivery to body tissues.32 Increased plasma levels of retinyl esters as a percentage of total vitamin A also occurred in human patients10,28,49 and rhesus monkeys41 from the same colony as those used in the current study. Numerous case studies in humans document various symptoms of hypervitaminosis A, including dermatologic, hepatic, and neurologic pathologies.27,30,35,47,48 Hepatic pathologies include abnormal liver function tests consistent with accumulation of lipid-storing droplets. Similar accumulation of lipid droplets has been reported to occur in rhesus monkeys from the same colony as those in the current study.39Previously, liver vitamin A concentrations in captive rhesus monkeys were reported to range from 11.9 ± 5.4 to 18.8 ± 6.4 μmol retinol/g liver,8,33,39 which are several fold higher than the concentrations considered excessive (that is, 0.70 to 1.05 μmol/g liver) and toxic (that is, 1.05 μmol/g liver) in humans.36 Hepatic vitamin A concentrations in 2 wild-caught control rhesus monkeys in a vitamin A deficiency study were 1.07 and 1.08 μmol retinol/g liver.38 Systematic inquiry to uncover the source of the high liver vitamin A concentrations found that the dietary vitamin A intake of captive rhesus monkeys exceeds National Research Council recommendations.40 Consistent with these excessive dietary vitamin A levels, clinical markers of hypervitaminosis A were present.8 To monitor the trafficking of a vitamin A dose during chronic hypervitaminosis A, we treated rhesus monkeys from the same colony as those cited earlier with 13C2-retinyl acetate and collected liver biopsies as part of a study reported elsewhere.8 The main goal of the previous report was to validate a vitamin A assessment technique using the heavy stable isotope of carbon.In the current study, we hypothesized that isotope ratio mass spectrometry (IRMS) could be used to detect accumulation of individual hepatic retinyl esters and provide evidence of de novo in vivo hepatic retinol esterification after treatment of rhesus macaques with a physiologic dose of 13C2-retinyl acetate. The 13C abundance of HPLC-purified hepatic retinyl esters collected at baseline was compared with that of posttreatment samples. Here we show that hepatic retinyl esters continue to accumulate in a nonhuman primate model by using state-of-the-art analytical methods and 13C-labeled retinol as a tracer.  相似文献   

2.
During a nearby construction project, a sudden decrease in food intake and guano production occurred in an outdoor colony of big brown bats (Eptesicus fuscus), and one animal was found dead. Investigation revealed that the project was generating a large amount of noise and vibration, which disturbed the bats’ feeding. Consequently the bats were moved into an indoor enclosure away from the construction noises, and the colony resumed eating. Over the next 3 wk, additional animals presented with clinical signs of lethargy, weight loss, ecchymoses, and icterus and were necropsied. Gross necropsy of the affected bats revealed large, pale yellow to tan, friable livers with rounded edges that floated when placed in 10% neutral-buffered formalin. Some bats had ecchymoses on the webbing and skin and gross perirenal hemorrhage. Histologic examination showed hepatic and renal tubular lipidosis. The clinical and pathologic signs of hemorrhage and icterus were suggestive of hepatic failure. Hepatic lipidosis was attributed to stress and inappetence associated with environmental perturbations. Once the environmental stressor was removed, the colony morbidity and mortality decreased. However, 2 y later, a series of new environmental stressors triggered additional deaths associated with hepatic lipidosis. Over a 9-y period, 21 cases of hepatic lipidosis were diagnosed in this bat colony.The big brown bat (Eptesicus fuscus), a member of the family Vespertilionidae, is an insectivorous species that relies on echolocation for the identification of prey and navigation during flight.1,2,19 Several subspecies with specific geographic distributions throughout the continental United States have been identified.6,24 Brown bats reach a typical adult weight of 13 to 25 g, give birth to 1 or 2 young per year, and hibernate in buildings, crevices, caves, and mines.19 Uses of this species in biomedical research include studies of spatial memory and flight dynamics, the integration of auditory stimuli through echolocation, and the development of echolocation and communication signals.1,21,23Spontaneously occurring medical problems in big brown bats include predation, trauma, emaciation, and infectious disease.3 Specific pathogens described in this species in the wild include white-nose syndrome (attributed to infection with Pseudogymnoascus destructans), Pasteurella multocida serotype 1, Demodex mites, rabies virus (Lyssavirus), and group 1 (Rocky Mountain bat) coronavirus.3,6,9,11,15,18 In addition, sporadic cases of cystitis with chronic interstitial nephritis and of hepatic lipidosis have been reported, although the etiology of hepatic lipidosis and concurrent clinical and pathologic findings were not discussed in these reports.2,18 Three captive horseshoe bats (Rhinolophus ferrumequinum) with hepatic, renal, and cardiac lipidosis, presumably related to stress and fat mobilization with excessive deposition of fatty acids in the liver and other organs, have been described.13We here describe a colony of big brown bats that developed lethargy, weight loss, and hemorrhage and were diagnosed with hepatic lipidosis due to exposure to an environmental stressor. The colony morbidity and mortality decreased after intervention; however, 2 y later, a series of new environmental stressors triggered additional deaths associated with hepatic lipidosis. Subsequent colony mortality was tracked, with infrequent cases of hepatic lipidosis occurring over the next 5 y.  相似文献   

3.
The DNA mismatch repair (MMR) system is a major DNA repair system that corrects DNA replication errors. In eukaryotes, the MMR system functions via mechanisms both dependent on and independent of exonuclease 1 (EXO1), an enzyme that has multiple roles in DNA metabolism. Although the mechanism of EXO1-dependent MMR is well understood, less is known about EXO1-independent MMR. Here, we provide genetic and biochemical evidence that the DNA2 nuclease/helicase has a role in EXO1-independent MMR. Biochemical reactions reconstituted with purified human proteins demonstrated that the nuclease activity of DNA2 promotes an EXO1-independent MMR reaction via a mismatch excision-independent mechanism that involves DNA polymerase δ. We show that DNA polymerase ε is not able to replace DNA polymerase δ in the DNA2-promoted MMR reaction. Unlike its nuclease activity, the helicase activity of DNA2 is dispensable for the ability of the protein to enhance the MMR reaction. Further examination established that DNA2 acts in the EXO1-independent MMR reaction by increasing the strand-displacement activity of DNA polymerase δ. These data reveal a mechanism for EXO1-independent mismatch repair.

The mismatch repair (MMR) system has been conserved from bacteria to humans (1, 2). It promotes genome stability by suppressing spontaneous and DNA damage-induced mutations (1, 3, 4, 5, 6, 7, 8, 9, 10, 11). The key function of the MMR system is the correction of DNA replication errors that escape the proofreading activities of replicative DNA polymerases (1, 4, 5, 6, 7, 8, 9, 10, 12). In addition, the MMR system removes mismatches formed during strand exchange in homologous recombination, suppresses homeologous recombination, initiates apoptosis in response to irreparable DNA damage caused by several anticancer drugs, and contributes to instability of triplet repeats and alternative DNA structures (1, 4, 5, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18). The principal components of the eukaryotic MMR system are MutSα (MSH2-MSH6 heterodimer), MutLα (MLH1-PMS2 heterodimer in humans and Mlh1-Pms1 heterodimer in yeast), MutSβ (MSH2-MSH3 heterodimer), proliferating cell nuclear antigen (PCNA), replication factor C (RFC), exonuclease 1 (EXO1), RPA, and DNA polymerase δ (Pol δ). Loss-of-function mutations in the MSH2, MLH1, MSH6, and PMS2 genes of the human MMR system cause Lynch and Turcot syndromes, and hypermethylation of the MLH1 promoter is responsible for ∼15% of sporadic cancers in several organs (19, 20). MMR deficiency leads to cancer initiation and progression via a multistage process that involves the inactivation of tumor suppressor genes and action of oncogenes (21).MMR occurs behind the replication fork (22, 23) and is a major determinant of the replication fidelity (24). The correction of DNA replication errors by the MMR system increases the replication fidelity by ∼100 fold (25). Strand breaks in leading and lagging strands as well as ribonucleotides in leading strands serve as signals that direct the eukaryotic MMR system to remove DNA replication errors (26, 27, 28, 29, 30). MMR is more efficient on the lagging than the leading strand (31). The substrates for MMR are all six base–base mismatches and 1 to 13-nt insertion/deletion loops (25, 32, 33, 34). Eukaryotic MMR commences with recognition of the mismatch by MutSα or MutSβ (32, 34, 35, 36). MutSα is the primary mismatch-recognition factor that recognizes both base–base mismatches and small insertion/deletion loops whereas MutSβ recognizes small insertion/deletion loops (32, 34, 35, 36, 37). After recognizing the mismatch, MutSα or MutSβ cooperates with RFC-loaded PCNA to activate MutLα endonuclease (38, 39, 40, 41, 42, 43). The activated MutLα endonuclease incises the discontinuous daughter strand 5′ and 3′ to the mismatch. A 5'' strand break formed by MutLα endonuclease is utilized by EXO1 to enter the DNA and excise a discontinuous strand portion encompassing the mismatch in a 5''→3′ excision reaction stimulated by MutSα/MutSβ (38, 44, 45). The generated gap is filled in by the Pol δ holoenzyme, and the nick is ligated by a DNA ligase (44, 46, 47). DNA polymerase ε (Pol ε) can substitute for Pol δ in the EXO1-dependent MMR reaction, but its activity in this reaction is much lower than that of Pol δ (48). Although MutLα endonuclease is essential for MMR in vivo, 5′ nick-dependent MMR reactions reconstituted in the presence of EXO1 are MutLα-independent (44, 47, 49).EXO1 deficiency in humans does not seem to cause significant cancer predisposition (19). Nevertheless, it is known that Exo1-/- mice are susceptible to the development of lymphomas (50). Genetic studies in yeast and mice demonstrated that EXO1 inactivation causes only a modest defect in MMR (50, 51, 52, 53). In agreement with these genetic studies, a defined human EXO1-independent MMR reaction that depends on the strand-displacement DNA synthesis activity of Pol δ holoenzyme to remove the mismatch was reconstituted (54). Furthermore, an EXO1-independent MMR reaction that occurred in a mammalian cell extract system without the formation of a gapped excision intermediate was observed (54). Together, these findings implicated the strand-displacement activity of Pol δ holoenzyme in EXO1-independent MMR.In this study, we investigated DNA2 in the context of MMR. DNA2 is an essential multifunctional protein that has nuclease, ATPase, and 5''→3′ helicase activities (55, 56, 57). Previous research ascertained that DNA2 removes long flaps during Okazaki fragment maturation (58, 59, 60), participates in the resection step of double-strand break repair (61, 62, 63), initiates the replication checkpoint (64), and suppresses the expansions of GAA repeats (65). We have found in vivo and in vitro evidence that DNA2 promotes EXO1-independent MMR. Our data have indicated that the nuclease activity of DNA2 enhances the strand-displacement activity of Pol δ holoenzyme in an EXO1-independent MMR reaction.  相似文献   

4.
A 5.5-y-old intact male cynomolgus macaque (Macaca fasicularis) presented with inappetence and weight loss 57 d after heterotopic heart and thymus transplantation while receiving an immunosuppressant regimen consisting of tacrolimus, mycophenolate mofetil, and methylprednisolone to prevent graft rejection. A serum chemistry panel, a glycated hemoglobin test, and urinalysis performed at presentation revealed elevated blood glucose and glycated hemoglobin (HbA1c) levels (727 mg/dL and 10.1%, respectively), glucosuria, and ketonuria. Diabetes mellitus was diagnosed, and insulin therapy was initiated immediately. The macaque was weaned off the immunosuppressive therapy as his clinical condition improved and stabilized. Approximately 74 d after discontinuation of the immunosuppressants, the blood glucose normalized, and the insulin therapy was stopped. The animal''s blood glucose and HbA1c values have remained within normal limits since this time. We suspect that our macaque experienced new-onset diabetes mellitus after transplantation, a condition that is commonly observed in human transplant patients but not well described in NHP. To our knowledge, this report represents the first documented case of new-onset diabetes mellitus after transplantation in a cynomolgus macaque.Abbreviations: NODAT, new-onset diabetes mellitus after transplantationNew-onset diabetes mellitus after transplantation (NODAT, formerly known as posttransplantation diabetes mellitus) is an important consequence of solid-organ transplantation in humans.7-10,15,17,19,21,25-28,31,33,34,37,38,42 A variety of risk factors have been identified including increased age, sex (male prevalence), elevated pretransplant fasting plasma glucose levels, and immunosuppressive therapy.7-10,15,17,19,21,25-28,31,33,34,37,38,42 The relationship between calcineurin inhibitors, such as tacrolimus and cyclosporin, and the development of NODAT is widely recognized in human medicine.7-10,15,17,19,21,25-28,31,33,34,37,38,42 Cynomolgus macaques (Macaca fasicularis) are a commonly used NHP model in organ transplantation research. Cases of natural and induced diabetes of cynomolgus monkeys have been described in the literature;14,43,45 however, NODAT in a macaque model of solid-organ transplantation has not been reported previously to our knowledge.  相似文献   

5.
6.
An aged male rhesus macaque in our colony had decreased appetite and a loss of interest in behavioral testing. CBC analysis revealed a regenerative, microcytic, hypochromic anemia with thrombocytosis, consistent with iron deficiency. A fecal occult blood test was positive. Ultrasound imaging revealed numerous, vascularized focal liver lesions that suggested metastases. The macaque''s appetite continued to decrease, and he became more lethargic. At this point, the investigator elected to euthanize the macaque. At necropsy, the ileocolic junction was white and abnormally thickened, and the liver was pale tan with approximately 18 discrete white masses randomly scattered throughout the hepatic parenchyma. Histologically, the mass at the ileocolic junction was identified as an intestinal adenocarcinoma, whereas the liver masses were confirmed to be undifferentiated hepatic sarcomas. This case report describes a rhesus macaque that had 2 unrelated primary neoplasms. A review of the literature indicates that this rhesus macaque is the first reported to have an adenocarcinoma of the ileocolic junction and multiple hepatic sarcomas simultaneously.Rhesus macaques (Macaca mulatta) are genetically similar to humans, have a similar aging phenotype at approximately 3 times the rate of those in humans, and develop spontaneous cancers similar to those in humans.36 In humans, gastrointestinal carcinomas are relatively common, but most of these lesions arise in the colon and rectum with only a small percentage in the small intestine and ileum.4,12,15,18 Although the ileocolic junction is considered a common site for intestinal adenocarcinomas in aged rhesus macaques, this tumor has also been found in the duodenum, jejunum, distal ileum, cecum, and colon.6,13,21-23,25,39 Intestinal adenocarcinomas also occur in aged cynomologus macaques (Macaca fasicularis),39 cotton-top marmosets (Saguinus oedipus),6,10 common marmosets (Callithrix jacchus),6,27 and a squirrel monkey (Saimiri sciureus).24 Cotton-top marmosets often develop adenocarcinomas of the colon, including the cecum–colon, and rectum.6,10 Common marmosets have been reported to develop adenocarcinomas of the small intestine.6,27 Adenocarcinoma of the cecum in a squirrel monkey has been reported.24Spontaneous hepatic tumors unrelated to carcinogenic factors, such as aflatoxin B1,33 occur only rarely in nonhuman primates. In the United States, primary malignant hepatic tumors in humans are rare, and fewer than 1% are reported to be hepatic sarcomas.1,16,40 Review of the nonhuman primate literature revealed reports of hepatic cholangiocarcinoma in a 25-y-old male capuchin monkey (Cebus albifrons),7 hepatocellular carcinoma in a 24-y-old male squirrel monkey (Saimiri boliviensis)5 and in a female squirrel monkey (Saimiri sciureus) older than 13 y,28 and hepatocellular carcinoma and cholangiocarcinoma in an African green monkey (Cercopithecus aethiops).34 Spontaneous hepatocellular carcinomas were reported to occur in 2 adolescent male cynomologus macaques younger than 5 y.31 Hepatic hemangiosarcoma was diagnosed in 3-y-old female rhesus macaque,26 and hepatic cholangiocarcinoma was found in a rhesus macaque that also had an intestinal adenocarcinoma.39The aged male rhesus macaque (Macaca mulatta) in the current case study was found to have adenocarcinoma of the ileocolic junction and multiple, random, discrete neoplasms in the liver, which were identified as undifferentiated sarcomas. No metastases from the intestinal adenocarcinoma were detected, but neoplastic cells similar to those of the undifferentiated hepatic cells were identified in an intestinal artery. The frequency of multiple tumor types in aged nonhuman primates is relevant to the use of older animals in research.  相似文献   

7.
The role of calcium-mediated signaling has been extensively studied in plant responses to abiotic stress signals. Calcineurin B-like proteins (CBLs) and CBL-interacting protein kinases (CIPKs) constitute a complex signaling network acting in diverse plant stress responses. Osmotic stress imposed by soil salinity and drought is a major abiotic stress that impedes plant growth and development and involves calcium-signaling processes. In this study, we report the functional analysis of CIPK21, an Arabidopsis (Arabidopsis thaliana) CBL-interacting protein kinase, ubiquitously expressed in plant tissues and up-regulated under multiple abiotic stress conditions. The growth of a loss-of-function mutant of CIPK21, cipk21, was hypersensitive to high salt and osmotic stress conditions. The calcium sensors CBL2 and CBL3 were found to physically interact with CIPK21 and target this kinase to the tonoplast. Moreover, preferential localization of CIPK21 to the tonoplast was detected under salt stress condition when coexpressed with CBL2 or CBL3. These findings suggest that CIPK21 mediates responses to salt stress condition in Arabidopsis, at least in part, by regulating ion and water homeostasis across the vacuolar membranes.Drought and salinity cause osmotic stress in plants and severely affect crop productivity throughout the world. Plants respond to osmotic stress by changing a number of cellular processes (Xiong et al., 1999; Xiong and Zhu, 2002; Bartels and Sunkar, 2005; Boudsocq and Lauriére, 2005). Some of these changes include activation of stress-responsive genes, regulation of membrane transport at both plasma membrane (PM) and vacuolar membrane (tonoplast) to maintain water and ionic homeostasis, and metabolic changes to produce compatible osmolytes such as Pro (Stewart and Lee, 1974; Krasensky and Jonak, 2012). It has been well established that a specific calcium (Ca2+) signature is generated in response to a particular environmental stimulus (Trewavas and Malhó, 1998; Scrase-Field and Knight, 2003; Luan, 2009; Kudla et al., 2010). The Ca2+ changes are primarily perceived by several Ca2+ sensors such as calmodulin (Reddy, 2001; Luan et al., 2002), Ca2+-dependent protein kinases (Harper and Harmon, 2005), calcineurin B-like proteins (CBLs; Luan et al., 2002; Batistič and Kudla, 2004; Pandey, 2008; Luan, 2009; Sanyal et al., 2015), and other Ca2+-binding proteins (Reddy, 2001; Shao et al., 2008) to initiate various cellular responses.Plant CBL-type Ca2+ sensors interact with and activate CBL-interacting protein kinases (CIPKs) that phosphorylate downstream components to transduce Ca2+ signals (Liu et al., 2000; Luan et al., 2002; Batistič and Kudla, 2004; Luan, 2009). In several plant species, multiple members have been identified in the CBL and CIPK family (Luan et al., 2002; Kolukisaoglu et al., 2004; Pandey, 2008; Batistič and Kudla, 2009; Weinl and Kudla, 2009; Pandey et al., 2014). Involvement of specific CBL-CIPK pair to decode a particular type of signal entails the alternative and selective complex formation leading to stimulus-response coupling (D’Angelo et al., 2006; Batistič et al., 2010).Several CBL and CIPK family members have been implicated in plant responses to drought, salinity, and osmotic stress based on genetic analysis of Arabidopsis (Arabidopsis thaliana) mutants (Zhu, 2002; Cheong et al., 2003, 2007; Kim et al., 2003; Pandey et al., 2004, 2008; D’Angelo et al., 2006; Qin et al., 2008; Tripathi et al., 2009; Held et al., 2011; Tang et al., 2012; Drerup et al., 2013; Eckert et al., 2014). A few CIPKs have also been functionally characterized by gain-of-function approach in crop plants such as rice (Oryza sativa), pea (Pisum sativum), and maize (Zea mays) and were found to be involved in osmotic stress responses (Mahajan et al., 2006; Xiang et al., 2007; Yang et al., 2008; Tripathi et al., 2009; Zhao et al., 2009; Cuéllar et al., 2010).In this report, we examined the role of the Arabidopsis CIPK21 gene in osmotic stress response by reverse genetic analysis. The loss-of-function mutant plants became hypersensitive to salt and mannitol stress conditions, suggesting that CIPK21 is involved in the regulation of osmotic stress response in Arabidopsis. These findings are further supported by an enhanced tonoplast targeting of the cytoplasmic CIPK21 through interaction with the vacuolar Ca2+ sensors CBL2 and CBL3 under salt stress condition.  相似文献   

8.
In the oxidative stress hypothesis of aging, the aging process is the result of cumulative damage by reactive oxygen species. Humans and chimpanzees are remarkably similar; but humans live twice as long as chimpanzees and therefore are believed to age at a slower rate. The purpose of this study was to compare biomarkers for cardiovascular disease, oxidative stress, and aging between male chimpanzees and humans. Compared with men, male chimpanzees were at increased risk for cardiovascular disease because of their significantly higher levels of fibrinogen, IGF1, insulin, lipoprotein a, and large high-density lipoproteins. Chimpanzees showed increased oxidative stress, measured as significantly higher levels of 5-hydroxymethyl-2-deoxyuridine and 8-iso-prostaglandin F, a higher peroxidizability index, and higher levels of the prooxidants ceruloplasmin and copper. In addition, chimpanzees had decreased levels of antioxidants, including α- and β-carotene, β-cryptoxanthin, lycopene, and tocopherols, as well as decreased levels of the cardiovascular protection factors albumin and bilirubin. As predicted by the oxidative stress hypothesis of aging, male chimpanzees exhibit higher levels of oxidative stress and a much higher risk for cardiovascular disease, particularly cardiomyopathy, compared with men of equivalent age. Given these results, we hypothesize that the longer lifespan of humans is at least in part the result of greater antioxidant capacity and lower risk of cardiovascular disease associated with lower oxidative stress.Abbreviations: 5OHmU, 5-hydroxymethyl-2-deoxyuridine; 8isoPGF, 8-iso-prostaglandin F; HDL, high-density lipoprotein; IGF1, insulin-like growth factor 1; LDL, low-density lipoprotein; ROS, reactive oxygen speciesAging is characterized as a progressive reduction in the capacity to withstand the stresses of everyday life and a corresponding increase in risk of mortality. According to the oxidative stress hypothesis of aging, much of the aging process can be accounted for as the result of cumulative damage produced by reactive oxygen species (ROS).6,21,28,41,97 Endogenous oxygen radicals (that is, ROS) are generated as a byproduct of normal metabolic reactions in the body and subsequently can cause extensive damage to proteins, lipids, and DNA.6,41 Various prooxidant elements, in particular free transition metals, can catalyze these destructive reactions.6 The damage caused by ROS can be counteracted by antioxidant defense systems, but the imbalance between production of ROS and antioxidant defenses, over time, leads to oxidative stress and may contribute to the rate of aging.28,97Oxidative stress has been linked to several age-related diseases including neurodegenerative diseases, ophthalmologic diseases, cancer, and cardiovascular disease.21,28,97 Of these, cardiovascular disease remains the leading cause of adult death in the United States and Europe.71 In terms of cardiovascular disease, oxidative stress has been linked to atherosclerosis, hypertension, cardiomyopathy, and chronic heart failure in humans.55,78,84 Increases in oxidant catalysts (prooxidants)—such as copper, iron, and cadmium—have been associated with hypertension, coronary artery disease, atherosclerosis, and sudden cardiac death.98,102,106 Finally, both endogenous and exogenous antioxidants have been linked to decreased risk of cardiovascular disease, although the mechanisms behind this relationship are unclear.11,52,53 However, the oxidative stress hypothesis of aging aims to explain not only the mechanism of aging and age-related diseases (such as cardiovascular disease) in humans but also the differences between aging rates and the manifestations of age-related diseases across species.The differences in antioxidant and ROS levels between animals and humans offer promise for increasing our understanding of human aging. Additional evidence supporting the oxidative stress hypothesis of aging has come from comparative studies linking differences in aging rates across taxa with both antioxidant and ROS levels.4,17-21,58,71,86,105 In mammals, maximum lifespan potential is positively correlated with both serum and tissue antioxidant levels.17,18,21,71,105 Research has consistently demonstrated that the rate of oxidative damage varies across species and is negatively correlated with maximum lifespan potential.4,19,20,58,71,86 However, few studies involved detailed comparisons of hypothesized biochemical indicators of aging and oxidative stress between humans and animals.6 This type of interspecies comparison has great potential for directly testing the oxidative stress hypothesis of aging.Much evolutionary and genetic evidence supports remarkable similarity between humans and chimpanzees.95,100 Despite this similarity, humans have a lifespan of almost twice that of chimpanzees.3,16,47 Most comparative primate aging research has focused on the use of a macaque model,62,81,88 and several biochemical markers of age-related diseases have been identified in both humans and macaque monkeys.9,22,28,81,93,97 Several other species of monkeys have also been used in research addressing oxidative stress, antioxidant defenses, and maximum lifespan potential.18,21,58,105 However, no study to date has examined biochemical indicators of oxidative stress and aging in chimpanzees and humans as a test of the oxidative stress hypothesis for aging. The purpose of this study is to compare biochemical markers for cardiovascular disease, oxidative stress, and aging directly between male chimpanzees and humans. Given the oxidative stress hypothesis for aging and the known role of oxidative stress in cardiovascular disease, we predict that chimpanzees will show higher levels of cardiovascular risk and oxidative stress than humans.  相似文献   

9.
Serum ceramides, especially C16:0 and C18:0 species, are linked to CVD risk and insulin resistance, but details of this association are not well understood. We performed this study to quantify a broad range of serum sphingolipids in individuals spanning the physiologic range of insulin sensitivity and to determine if dihydroceramides cause insulin resistance in vitro. As expected, we found that serum triglycerides were significantly greater in individuals with obesity and T2D compared with athletes and lean individuals. Serum ceramides were not significantly different within groups but, using all ceramide data relative to insulin sensitivity as a continuous variable, we observed significant inverse relationships between C18:0, C20:0, and C22:0 species and insulin sensitivity. Interestingly, we found that total serum dihydroceramides and individual species were significantly greater in individuals with obesity and T2D compared with athletes and lean individuals, with C18:0 species showing the strongest inverse relationship to insulin sensitivity. Finally, we administered a physiological mix of dihydroceramides to primary myotubes and found decreased insulin sensitivity in vitro without changing the overall intracellular sphingolipid content, suggesting a direct effect on insulin resistance. These data extend what is known regarding serum sphingolipids and insulin resistance and show the importance of serum dihydroceramides to predict and promote insulin resistance in humans.Supplementary key words: sphingolipids, circulating ceramides, serum, insulin resistance, lipidomics, CVD, T2D, obesity, myotube

Circulating ceramides, especially specific saturated ceramide species, and other sphingolipids are linked to CVD risk and insulin resistance (1, 2, 3, 4, 5, 6, 7, 8, 9, 10). In fact, circulating ceramide and sphingolipid contents predict development of CVD better than some common risk factors such as plasma cholesterol, LDLs, and triglycerides (6, 9, 11, 12). As a result, it was recently proposed that plasma ceramide could be the new cholesterol for assessing risk of CVD (11). Beyond the cross-sectional studies referenced above, there are several lines of evidence supporting the link between ceramides, CVD, and insulin resistance. Plasma ceramide content decreases after insulin-sensitizing gastric bypass surgery and weight loss interventions (13, 14, 15). Animal studies show that ceramides accumulate in atherosclerotic lesions, which may explain the increased risk associated with plasma content (16). However, the relationship of circulating sphingolipids to insulin resistance is not absolute, as insulin-sensitizing treatments do not always change plasma sphingolipid content (17). Combined, most data from epidemiology studies, as well as human interventions and animal models, support the concept that circulating ceramides and sphingolipids are related to insulin resistance and CVD risk.Ceramides circulate primarily bound to lipoproteins and are secreted predominately by the liver. Circulating ceramides are mainly increased in LDL in individuals with obesity (15). Obese rodents have increased hepatic ceramide secretion, which may explain increased plasma ceramide content in individuals with obesity (15). In one mechanistic study, an LDL-ceramide mixture was infused in mice to recapitulate increased plasma ceramide content in obesity, which caused membrane ceramide accumulation, decreased insulin signaling, and a decrease in insulin sensitivity specifically in skeletal muscle, providing evidence for a direct effect of circulating ceramides on tissues (15). Similarly, LDL-ceramide administration to myotubes caused ceramide accumulation, decreased insulin sensitivity, and signaling independent of inflammation. These data indicate that plasma ceramides are not simply markers of insulin resistance but play mechanistic roles in decreasing insulin sensitivity.Ceramides are only one member of the sphingolipid family, and other sphingolipids may also be related to insulin resistance and CVD risk. Lactosylceramides and glucosylceramides are sphingolipids that also accumulate in atherosclerotic plaques and therefore may be involved in the CVD process (18). Sphingomyelins are the most abundant sphingolipids circulating in lipoproteins and, while they are positively related to obesity and waist circumference, they are not correlated to insulin sensitivity in cross-sectional human studies (5, 19). Dihydroceramides are immediate precursors to ceramide synthesis and are negatively related to insulin sensitivity (20, 21) and insulin secretion (21), are positively related to waist circumference (22), are elevated in plasma of individuals with prediabetes and T2D compared with controls (23), and predict development of diabetes 9 years before onset (21). Despite strong evidence linking plasma dihydroceramides to decreased insulin sensitivity, mechanistic studies to determine if circulating dihydroceramides cause insulin resistance are lacking.To address this knowledge gap, we performed the current study to assess serum sphingolipids in humans across the metabolic spectrum as well as determine if dihydroceramides induce insulin resistance in vitro.  相似文献   

10.
11.
12.
Tumor necrosis factor α (TNFα) triggers necroptotic cell death through an intracellular signaling complex containing receptor-interacting protein kinase (RIPK) 1 and RIPK3, called the necrosome. RIPK1 phosphorylates RIPK3, which phosphorylates the pseudokinase mixed lineage kinase-domain-like (MLKL)—driving its oligomerization and membrane-disrupting necroptotic activity. Here, we show that TNF receptor-associated factor 2 (TRAF2)—previously implicated in apoptosis suppression—also inhibits necroptotic signaling by TNFα. TRAF2 disruption in mouse fibroblasts augmented TNFα–driven necrosome formation and RIPK3-MLKL association, promoting necroptosis. TRAF2 constitutively associated with MLKL, whereas TNFα reversed this via cylindromatosis-dependent TRAF2 deubiquitination. Ectopic interaction of TRAF2 and MLKL required the C-terminal portion but not the N-terminal, RING, or CIM region of TRAF2. Induced TRAF2 knockout (KO) in adult mice caused rapid lethality, in conjunction with increased hepatic necrosome assembly. By contrast, TRAF2 KO on a RIPK3 KO background caused delayed mortality, in concert with elevated intestinal caspase-8 protein and activity. Combined injection of TNFR1-Fc, Fas-Fc and DR5-Fc decoys prevented death upon TRAF2 KO. However, Fas-Fc and DR5-Fc were ineffective, whereas TNFR1-Fc and interferon α receptor (IFNAR1)-Fc were partially protective against lethality upon combined TRAF2 and RIPK3 KO. These results identify TRAF2 as an important biological suppressor of necroptosis in vitro and in vivo.Apoptotic cell death is mediated by caspases and has distinct morphological features, including membrane blebbing, cell shrinkage and nuclear fragmentation.1, 2, 3, 4 In contrast, necroptotic cell death is caspase-independent and is characterized by loss of membrane integrity, cell swelling and implosion.1, 2, 5 Nevertheless, necroptosis is a highly regulated process, requiring activation of RIPK1 and RIPK3, which form the core necrosome complex.1, 2, 5 Necrosome assembly can be induced via specific death receptors or toll-like receptors, among other modules.6, 7, 8, 9 The activated necrosome engages MLKL by RIPK3-mediated phosphorylation.6, 10, 11 MLKL then oligomerizes and binds to membrane phospholipids, forming pores that cause necroptotic cell death.10, 12, 13, 14, 15 Unchecked necroptosis disrupts embryonic development in mice and contributes to several human diseases.7, 8, 16, 17, 18, 19, 20, 21, 22The apoptotic mediators FADD, caspase-8 and cFLIP suppress necroptosis.19, 20, 21, 23, 24 Elimination of any of these genes in mice causes embryonic lethality, subverted by additional deletion of RIPK3 or MLKL.19, 20, 21, 25 Necroptosis is also regulated at the level of RIPK1. Whereas TNFα engagement of TNFR1 leads to K63-linked ubiquitination of RIPK1 by cellular inhibitor of apoptosis proteins (cIAPs) to promote nuclear factor (NF)-κB activation,26 necroptosis requires suppression or reversal of this modification to allow RIPK1 autophosphorylation and consequent RIPK3 activation.2, 23, 27, 28 CYLD promotes necroptotic signaling by deubiquitinating RIPK1, augmenting its interaction with RIPK3.29 Conversely, caspase-8-mediated CYLD cleavage inhibits necroptosis.24TRAF2 recruits cIAPs to the TNFα-TNFR1 signaling complex, facilitating NF-κB activation.30, 31, 32, 33 TRAF2 also supports K48-linked ubiquitination and proteasomal degradation of death-receptor-activated caspase-8, curbing apoptosis.34 TRAF2 KO mice display embryonic lethality; some survive through birth but have severe developmental and immune deficiencies and die prematurely.35, 36 Conditional TRAF2 KO leads to rapid intestinal inflammation and mortality.37 Furthermore, hepatic TRAF2 depletion augments apoptosis activation via Fas/CD95.34 TRAF2 attenuates necroptosis induction in vitro by the death ligands Apo2L/TRAIL and Fas/CD95L.38 However, it remains unclear whether TRAF2 regulates TNFα-induced necroptosis—and if so—how. Our present findings reveal that TRAF2 inhibits TNFα necroptotic signaling. Furthermore, our results establish TRAF2 as a biologically important necroptosis suppressor in vitro and in vivo and provide initial insight into the mechanisms underlying this function.  相似文献   

13.
Neuropeptides induce signal transduction across the plasma membrane by acting through cell-surface receptors. The dynorphins, endogenous ligands for opioid receptors, are an exception; they also produce non-receptor-mediated effects causing pain and neurodegeneration. To understand non-receptor mechanism(s), we examined interactions of dynorphins with plasma membrane. Using fluorescence correlation spectroscopy and patch-clamp electrophysiology, we demonstrate that dynorphins accumulate in the membrane and induce a continuum of transient increases in ionic conductance. This phenomenon is consistent with stochastic formation of giant (~2.7 nm estimated diameter) unstructured non-ion-selective membrane pores. The potency of dynorphins to porate the plasma membrane correlates with their pathogenic effects in cellular and animal models. Membrane poration by dynorphins may represent a mechanism of pathological signal transduction. Persistent neuronal excitation by this mechanism may lead to profound neuropathological alterations, including neurodegeneration and cell death.Neuropeptides are the largest and most diverse family of neurotransmitters. They are released from axon terminals and dendrites, diffuse to pre- or postsynaptic neuronal structures and activate membrane G-protein-coupled receptors. Prodynorphin (PDYN)-derived opioid peptides including dynorphin A (Dyn A), dynorphin B (Dyn B) and big dynorphin (Big Dyn) consisting of Dyn A and Dyn B are endogenous ligands for the κ-opioid receptor. Acting through this receptor, dynorphins regulate processing of pain and emotions, memory acquisition and modulate reward induced by addictive substances.1, 2, 3, 4 Furthermore, dynorphins may produce robust cellular and behavioral effects that are not mediated through opioid receptors.5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 As evident from pharmacological, morphological, genetic and human neuropathological studies, these effects are generally pathological, including cell death, neurodegeneration, neurological dysfunctions and chronic pain. Big Dyn is the most active pathogenic peptide, which is about 10- to 100-fold more potent than Dyn A, whereas Dyn B does not produce non-opioid effects.16, 17, 22, 25 Big Dyn enhances activity of acid-sensing ion channel-1a (ASIC1a) and potentiates ASIC1a-mediated cell death in nanomolar concentrations30, 31 and, when administered intrathecally, induces characteristic nociceptive behavior at femtomolar doses.17, 22 Inhibition of endogenous Big Dyn degradation results in pathological pain, whereas prodynorphin (Pdyn) knockout mice do not maintain neuropathic pain.22, 32 Big Dyn differs from its constituents Dyn A and Dyn B in its unique pattern of non-opioid memory-enhancing, locomotor- and anxiolytic-like effects.25Pathological role of dynorphins is emphasized by the identification of PDYN missense mutations that cause profound neurodegeneration in the human brain underlying the SCA23 (spinocerebellar ataxia type 23), a very rare dominantly inherited neurodegenerative disorder.27, 33 Most PDYN mutations are located in the Big Dyn domain, demonstrating its critical role in neurodegeneration. PDYN mutations result in marked elevation in dynorphin levels and increase in its pathogenic non-opioid activity.27, 34 Dominant-negative pathogenic effects of dynorphins are not produced through opioid receptors.ASIC1a, glutamate NMDA (N-methyl-d-aspartate) and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid)/kainate ion channels, and melanocortin and bradykinin B2 receptors have all been implicated as non-opioid dynorphin targets.5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 30, 31, 35, 36 Multiplicity of these targets and their association with the cellular membrane suggest that their activation is a secondary event triggered by a primary interaction of dynorphins with the membrane. Dynorphins are among the most basic neuropeptides.37, 38 The basic nature is also a general property of anti-microbial peptides (AMPs) and amyloid peptides that act by inducing membrane perturbations, altering membrane curvature and causing pore formation that disrupts membrane-associated processes including ion fluxes across the membrane.39 The similarity between dynorphins and these two peptide groups in overall charge and size suggests a similar mode of their interactions with membranes.In this study, we dissect the interactions of dynorphins with the cell membrane, the primary event in their non-receptor actions. Using fluorescence imaging, correlation spectroscopy and patch-clamp techniques, we demonstrate that dynorphin peptides accumulate in the plasma membrane in live cells and cause a profound transient increase in cell membrane conductance. Membrane poration by endogenous neuropeptides may represent a novel mechanism of signal transduction in the brain. This mechanism may underlie effects of dynorphins under pathological conditions including chronic pain and tissue injury.  相似文献   

14.
Necroptosis is a form of regulated necrotic cell death mediated by receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3. Necroptotic cell death contributes to the pathophysiology of several disorders involving tissue damage, including myocardial infarction, stroke and ischemia-reperfusion injury. However, no inhibitors of necroptosis are currently in clinical use. Here we performed a phenotypic screen for small-molecule inhibitors of tumor necrosis factor-alpha (TNF)-induced necroptosis in Fas-associated protein with death domain (FADD)-deficient Jurkat cells using a representative panel of Food and Drug Administration (FDA)-approved drugs. We identified two anti-cancer agents, ponatinib and pazopanib, as submicromolar inhibitors of necroptosis. Both compounds inhibited necroptotic cell death induced by various cell death receptor ligands in human cells, while not protecting from apoptosis. Ponatinib and pazopanib abrogated phosphorylation of mixed lineage kinase domain-like protein (MLKL) upon TNF-α-induced necroptosis, indicating that both agents target a component upstream of MLKL. An unbiased chemical proteomic approach determined the cellular target spectrum of ponatinib, revealing key members of the necroptosis signaling pathway. We validated RIPK1, RIPK3 and transforming growth factor-β-activated kinase 1 (TAK1) as novel, direct targets of ponatinib by using competitive binding, cellular thermal shift and recombinant kinase assays. Ponatinib inhibited both RIPK1 and RIPK3, while pazopanib preferentially targeted RIPK1. The identification of the FDA-approved drugs ponatinib and pazopanib as cellular inhibitors of necroptosis highlights them as potentially interesting for the treatment of pathologies caused or aggravated by necroptotic cell death.Programmed cell death has a crucial role in a variety of biological processes ranging from normal tissue development to diverse pathological conditions.1, 2 Necroptosis is a form of regulated cell death that has been shown to occur during pathogen infection or sterile injury-induced inflammation in conditions where apoptosis signaling is compromised.3, 4, 5, 6 Given that many viruses have developed strategies to circumvent apoptotic cell death, necroptosis constitutes an important, pro-inflammatory back-up mechanism that limits viral spread in vivo.7, 8, 9 In contrast, in the context of sterile inflammation, necroptotic cell death contributes to disease pathology, outlining potential benefits of therapeutic intervention.10 Necroptosis can be initiated by death receptors of the tumor necrosis factor (TNF) superfamily,11 Toll-like receptor 3 (TLR3),12 TLR4,13 DNA-dependent activator of IFN-regulatory factors14 or interferon receptors.15 Downstream signaling is subsequently conveyed via RIPK116 or TIR-domain-containing adapter-inducing interferon-β,8, 17 and converges on RIPK3-mediated13, 18, 19, 20 activation of MLKL.21 Phosphorylated MLKL triggers membrane rupture,22, 23, 24, 25, 26 releasing pro-inflammatory cellular contents to the extracellular space.27 Studies using the RIPK1 inhibitor necrostatin-1 (Nec-1) 28 or RIPK3-deficient mice have established a role for necroptosis in the pathophysiology of pancreatitis,19 artherosclerosis,29 retinal cell death,30 ischemic organ damage and ischemia-reperfusion injury in both the kidney31 and the heart.32 Moreover, allografts from RIPK3-deficient mice are better protected from rejection, suggesting necroptosis inhibition as a therapeutic option to improve transplant outcome.33 Besides Nec-1, several tool compounds inhibiting different pathway members have been described,12, 16, 21, 34, 35 however, no inhibitors of necroptosis are available for clinical use so far.2, 10 In this study we screened a library of FDA approved drugs for the precise purpose of identifying already existing and generally safe chemical agents that could be used as necroptosis inhibitors. We identified the two structurally distinct kinase inhibitors pazopanib and ponatinib as potent blockers of necroptosis targeting the key enzymes RIPK1/3.  相似文献   

15.
Retinoic acid is a widely used factor in both mouse and human embryonic stem cells. It suppresses differentiation to mesoderm and enhances differentiation to ectoderm. Fibroblast growth factor 2 (FGF2) is widely used to induce differentiation to neurons in mice, yet in primates, including humans, it maintains embryonic stem cells in the undifferentiated state. In this study, we established an FGF2 low-dose-dependent embryonic stem cell line from cynomolgus monkeys and then analyzed neural differentiation in cultures supplemented with retinoic acid and FGF2. When only retinoic acid was added to culture, neurons differentiated from FGF2 low-dose-dependent embryonic stem cells. When both retinoic acid and FGF2 were added, neurons and astrocytes differentiated from the same embryonic stem cell line. Thus, retinoic acid promotes the differentiation from embryonic stem cells to neuroectoderm. Although FGF2 seems to promote self-renewal in stem cells, its effects on the differentiation of stem cells are influenced by the presence or absence of supplemental retinoic acid.Abbreviations: EB, embryoid body; ES, embryonic stem; ESM, embryonic stem cell medium; FGF, fibroblast growth factor; GFAP, glial fibrillary acidic protein; LIF, leukemia inhibitory factor; MBP, myelin basic protein; RA, retinoic acid; SSEA, stage-specific embryonic antigen; TRA, tumor-related antigenPluripotent stem cells are potential sources of material for cell replacement therapy and are useful experimental tools for in vitro models of human disease and drug screening. Embryonic stem (ES) cells are capable of extensive proliferation and multilineage differentiation, and thus ES-derived cells are suitable for use in cell-replacement therapies.18,23 Reported ES cell characteristics including tumorigenic potential, DNA methylation status, expression of imprinted genes, and chromatin structure were elucidated by using induced pluripotent stem cells.2,11,17 Because the social expectations of regeneration medicine are growing, we must perform basic research with ES cells, which differ from induced pluripotent stem cells in terms of origin, differentiation ability, and epigenetic status.2,8Several advances in research have been made by using mouse ES cells. Furthermore, primate ES cell lines have been established from rhesus monkeys (Macaca mulatta),24 common marmosets (Callithrix jacchus),25 cynomolgus monkeys (M. fascicularis),20 and African green monkeys (Chlorocebus aethiops).19 Mouse and other mammalian ES cells differ markedly in their responses to the signaling pathways that support self-renewal.8,28 Mouse ES cells require leukemia inhibitory factor (LIF)–STAT3 signaling.14 In contrast, primate ES cells do not respond to LIF. Fibroblast growth factor 2 (FGF2) appears to be the most upstream self-renewal factor in primate ES cells. FGF2 also exerts its effects through indirect mechanisms, such as the TGFβ–Activin–Nodal signaling pathway, in primate ES cells.21 In addition to the biologic similarities between monkeys and humans, ES cells derived from cynomolgus monkeys or human blastocysts have extensive similarities that are not apparent in mouse ES cells.8,14,21,28 Numerous monkey ES cell lines are now available, and cynomolgus monkeys are an efficient model for developing strategies to investigate the efficacy of ES-cell–based medical treatments in humans.Several growth factors and chemical compounds, including retinoic acid (RA),4,9,13,22,26 FGF2,9,10,16,22 epidermal growth factor,9,22 SB431542,1,4,10 dorsomorphin,10,27 sonic hedgehog,12,13,16,27,29 and noggin,1,4,9,27 are essential for the differentiation and proliferation or maintenance of neural stem cells derived from primate ES cells. Of these factors, active RA signaling suppresses a mesodermal fate by inhibiting Wnt and Nodal signaling pathways during in vitro culture and leads to neuroectoderm differentiation in ES cells.4,13,26 RA is an indispensable factor for the specialization to neural cells. FGF2 is important during nervous system development,12 and FGF2 and RA both are believed to influence the differentiation to neural cells. The current study was done to clarify the mechanism of RA and FGF2 in the induction of differentiation along the neural lineage.We recently established a monkey ES cell line that does not need FGF2 supplementation for maintenance of the undifferentiated state. This ES cell line allowed us to study the role of differentiation to neural cells with RA and enabled us to compare ES cell differentiation in the context of supplementation with RA or FGF2 in culture. To this end, we established a novel cynomolgus monkey cell line derived from ES cells and maintained it in an undifferentiated state in the absence of FGF2 supplementation.  相似文献   

16.
17.
A retrospective study using maternal and birth statistics from an open, captive rhesus macaque colony was done to determine the effects of parity, exposure to simian retrovirus (SRV), housing, maternal parity, and maternal birth weight on infant birth weight, viability and gestation length. Retrospective colony statistics for a 23-y period indicated that birth weight, but not gestation length, differed between genders. Adjusted mean birth weights were higher in nonviable infants. Mothers positive for SRV had shorter gestations, but SRV exposure did not affect neonatal birth weights or viability. Infants born in cages had longer gestations than did those born in pens, but neither birth weight nor viability differed between these groups. Maternal birth weight did not correlate with infant birth weight but positively correlated with gestation length. Parity was correlated with birth weight and decreased viability. Increased parity of the mother was associated with higher birth weight of the infant. A transgenerational trend toward increasing birth weight was noted. The birth statistics of this colony were consistent with those of other macaque colonies. Unlike findings for humans, maternal birth weight had little predictive value for infant outcomes in rhesus macaques. Nonviable rhesus infants had higher birth weights, unlike their human counterparts, perhaps due to gestational diabetes occurring in a sedentary caged population. Similar to the situation for humans, multiparity had a protective effect on infant viability in rhesus macaques.Abbreviations: ANCOVA, analysis of covariance; PRL, Primate Research Laboratory; SRV, simian retrovirusThe rhesus macaque (Macaca mulatta) is a useful animal model for human female reproduction studies because the comparative physiology between the 2 species is nearly identical.1.5,49 Some factors that affect birth weight and neonatal viability in both humans and macaques include maternal birth weight, maternal age, maternal parity, and the presence of underlying maternal disease. Even experimentally induced simulated human lifestyle factors can affect neonatal outcome.10,16,17,25,44In humans, maternal birth weight correlates with infant birth weight such that low birth weight mothers themselves have low birth weight infants.8,19,28,30 A similar association has been shown in the macaque.38,39 Because low birth weight is associated with increased neonatal mortality in humans and in macaques, this correlation, if present, may have important predictive value.11,20,21,32,45,47,53 One objective of this study was to establish whether maternal birth weight correlated with neonatal birth weight and viability in this colony of rhesus macaques.The relationship between parity, age, and birth outcomes in humans is controversial because multiparous and grand multiparous women tend to be of lower socioeconomic status, older, and have many confounding lifestyle factors.2,24,27,56 In macaques, low parity and young age are associated with reproductive failure.50 In pigtailed macaques (Macaca nemestrina), increased parity was associated with decreased neonatal viability but increased birth weight. Despite their lower parity, younger mothers in the colony of pigtailed macaques produced lower birth-weight infants, but more viable infants, compared with those of older mothers.17 The positive correlation between birth weight and viability merits further investigation in rhesus macaques. One objective of the current study was to determine whether maternal parity and age affected birth weight and neonatal viability in our rhesus macaque colony.The lifestyle factors of alcohol consumption, cigarettes, caffeine, drug use, diabetes and exercise have all been shown to influence birth weight and gestation length in humans and macaques.4,7,15,22,26,35,40,42,44,51,55 Captive animals can become obese and develop insulin-resistant diabetes, which prolongs gestation and produces oversized infants that are less healthy.21,46,51 Because exercise is a preventative lifestyle factor for obesity and diabetes, it would be useful to compare active animals with sedentary ones.30 Previous retrospective colony studies in pigtail macaques show that cage type, location, and social housing have significant effects on birth weight and birth outcome.18,19 Another objective of the current study was to determine whether housing in cages (sedentary animals) or group pens (active animals) influenced gestation length, birth weight, and viability in our rhesus macaques.Another factor in birth outcome is the disease status of the mother. Viral infections, particularly of adenoviruses and immunosuppressive retroviruses, are associated with low birth weight and infant mortality in humans and nonhuman primates.13,21,25,33, 34,52,53 A previous report describes maternal transmission of simian retrovirus in a colony of pigtailed macaques with concurrent immunosuppression, low birth weight, and increased infant mortality in viremic mothers.33 However, some evidence suggests that lentiviral antibodies in amniotic fluid may protect against in utero infection.23 Further confounding the effects of retroviruses on reproductive outcome, animals infected horizontally can be viremic but serologically negative, and animals with sufficient, detectable immune responses may have provirus latent in their tissues.33 Because simian retrovirus (SRV) was endemic in the subject rhesus colony and most data were retrospective thus preventing confirmation of viremia, another objective was to determine whether seropositivity of the dam was associated with neonatal viability, gestation length, and infant birth weight.  相似文献   

18.
19.
Ulcerative dermatitis (UD) is a common condition in C57BL/6 mice and strains with this background. The etiology of UD is unclear but appears to have a genetic component associated with the C57BL/6 strain and has been reported as secondary to a variety of conditions. Treatment is unrewarding, resulting in euthanasia in many cases. In the present study we compared 3 topical treatments against spontaneous UD in mice with a C57BL/6 background. In total, 301 mice of both sexes were included in this study, and the tested treatments comprised bacitracin–neomycin sulfate–polymixin B sulfate ointment twice daily, 10% povidone–iodine ointment plus 1% silver sulfadiazine cream once daily, and 0.005% sodium hypochlorite once daily. Lesion healing was defined as complete skin reepithelialization with or without hair regrowth. Sex, age, lesion location, and type and length of treatment were analyzed by using univariate and multivariate logistic regression. Of the 79 mice treated with triple-antibiotic ointment, 27 (34%) healed, compared with 43 of the 125 (34%) treated with povidone–iodine and sulfadiazine and 69 of the 97 (71%) treated with hypochlorite. Lesion size and treatment with 0.005% sodium hypochlorite were the only significant predictors of healing; all other variables were not statistically significant in multivariate analysis. We conclude that 0.005% sodium hypochlorite is an effective topical treatment alternative for UD in C57BL/6 mice and strains on this background, and a favorable prognosis depends on the early identification and treatment of those lesions.Abbreviations: B6, C57BL/6; UD, ulcerative dermatitisUlcerative dermatitis (UD) is a common condition in C57BL/6 (B6) mice and strains with a B6 background.1,21 Early lesions are characterized by small skin erosions that can affect any part of the body but are typically found between the scapulae. Usually these lesions rapidly progress to form large, irregular areas of ulcerated skin.1 The condition can be very pruritic, resulting in self-mutilation, skin degloving, and exposure of the subcutaneous tissues and, in some cases, musculature.1 Common sequelae in mice that recover from this disease are marked lymphadenopathy and splenomegaly due to reactive immune modulation or activation, which can confound research results.21,31 When UD affects extensive areas and then heals, contracture and scarring of the skin cause tension that alters normal posture and ambulation.1Primary (idiopathic) UD is diagnosed by ruling out other conditions that cause dermatitis (secondary) in laboratory mice, such as allergy to fur mites,8,18 fight wounds, staphylococcal skin infections,20,32 phenotype,19,21,31 and experimental manipulation.13,22,33 The exact etiology of UD remains undetermined but seems to be multifactorial.9 Proposed etiologies include behavioral,10,11,34,35 immune-complex–induced vasculitis,1 cellular oxidative injury,21 and vitamin A toxicity.31 Calorie-restricted diets, providing 60% of the average calorie intake of the respective unrestricted group, seem to reduce ulcerative dermatitis,28 whereas high-fat diets (35% crude fat) appear to exacerbate the condition.27 UD has been reported to affect more female than male mice, with the highest incidence in mice older than 1 y, but UD can also occur in young mice.1,19,31 Although UD occurs throughout the year, some authors report a peak incidence during spring and fall, whereas others note increased case numbers during the summer months.19,31Attempts to find a cure for UD have not found a treatment that is completely effective. Treatment typically is unrewarding, resulting in euthanasia in many cases.21 Dietary supplementation with vitamin E reportedly has some efficacy favoring skin reepithelization in mice with UD.21 However, a recent study using vitamin E as a diet supplement to prevent the occurrence of UD yielded contradictory results.24 In that study, mice fed a vitamin-E–fortified diet since weaning were more likely to develop UD than were mice fed a regular diet. However, to achieve the desire amount of vitamin E, the fat content of the diet had to be increased; high dietary fat is known to exacerbate UD.24,27 Other studies have shown systemic administration of maropitant citrate reduces the size of UD lesions in mice by decreasing scratching,35 and the oral administration of ibuprofen appears to help speed the healing of skin lesions by reducing inflammation and pain.11 Topical and systemic antibiotics, corticosteroids, antihistamines, and lidocaine are poorly effective in the treatment of UD.1,19,21,31 Among topical treatments, caladryl lotion, chlorhexidine, and cyclosporine appear to be the most effective in treating UD.7,12,23 Toenail trimming has been reported as effective at reducing self-trauma due to scratching in UD, thus helping to speed healing.26,29In the present study, we compared 3 topical treatments against spontaneous UD in mice with a B6 background.  相似文献   

20.
Biology and tropism of MuAstV2Murine astrovirus 2 (MuAstV2) is a novel murine astrovirus recently identified in laboratory and wild mice. MuAstV2 readily transmits between immunocompetent mice yet fails to transmit to highly immunocompromised mouse strains—a unique characteristic when contrasted with other murine viruses including other astroviruses. We characterized the viral shedding kinetics and tissue tropism of MuAstV2 in immunocompetent C57BL/6NCrl mice and evaluated the apparent resistance of highly immunocompromised NOD-Prkdcem26Cd52Il2rgem26Cd22/NjuCrl mice to MuAstV2 after oral inoculation. Temporal patterns of viral shedding were determined by serially measuring fecal viral RNA. Tissue tropism and viral load were characterized and quantified by using in-situ hybridization (ISH) targeting viral RNA. Cellular tropism was characterized by evaluating fluorescent colocalization of viral ISH with various immunohistochemical markers. We found a rapid increase of fecal viral RNA in B6 mice, which peaked at 5 d after inoculation (dpi) followed by cessation of shedding by 168 dpi. The small intestine had the highest percentage of hybridization (3.09% of tissue area) of all tissues in which hybridization occurred at 5 dpi. The thymus displayed the next highest degree of hybridization (2.3%) at 7 dpi, indicating extraintestinal viral spread. MuAstV2 RNA hybridization was found to colocalize with only 3 of the markers evaluated: CD3 (T cells), Iba1 (macrophages), and cytokeratin (enterocytes). A higher percentage of CD3 cells and Iba1 cells hybridized with MuAstV2 as compared with cytokeratin at 2 dpi (CD3, 59%; Iba1, 46%; cytokeratin, 6%) and 35 dpi (CD3, 14%; Iba1, 55%; cytokeratin, 3%). Neither fecal viral RNA nor viral hybridization was noted in NCG mice at the time points examined. In addition, mice of mixed genetic background were inoculated, and only those with a functioning Il2rg gene shed MuAstV2. Results from this study suggest that infection of, or interaction with, the immune system is required for infection by or replication of MuAstV2.

Astroviruses are nonenveloped, positive-sense, single-stranded RNA viruses with a star-like appearance—from which the name derives—when examined by transmission electron microscopy. First identified in 1975, astroviruses are commonly associated with gastrointestinal illness in children.1,23 They demonstrate considerable diversity, and unique strains have been identified in numerous species through advances in molecular diagnostics.2,4-6,11-13,18,19,21,25,27,29,30,32,35-40 This broad distribution likely resulted from cross-species transmission and subsequent adaptation to the novel host.11 Clinical presentation varies among species, although most infections are asymptomatic or limited to mild gastrointestinal illness.4,9,11 Extraintestinal disease resulting in fatal encephalitis has been described in several species (including cows, mink, and immunocompromised people).3,19,22,26,35Astroviral infection of mice was first described in 1985, when an unknown astrovirus was identified by electron microscopy in the feces of nude mice.17 Since then, astroviruses have been detected in many wild and laboratory mouse populations.12,29,30,34 Despite their prevalence, studies have been limited and their effects on host biology remains largely unknown. Murine astrovirus (MuAstV) was identified through molecular sequencing in 2012 and has since been discovered to be enzootic in numerous research and production mouse colonies.12,29,34 Whether the strain described in 1985 was MuAstV is unknown. Immunocompetent and immunodeficient mouse strains are both susceptible to MuAstV infection, although no clinical disease and only minimal pathology are observed.7,47 Similar to astroviruses infecting other species, MuAstV infection is frequently localized to the gastrointestinal tract.47 A recent study demonstrated MuAstV replication in goblet cells and altered mucus production within the gastrointestinal tract, highlighting the potential effect of the virus on select research studies despite the lack of clinical disease and pathology.8Our group previously reported the detection of a novel murine astrovirus, murine astrovirus 2 (MuAstV2), in a laboratory mouse colony.31 MuAstV2 is genetically distinct from MuAstV but is closely related to a strain recently reported in wild mice.31,43 The MuAstV2 strain identified in the laboratory mouse colony shares 89.2% nucleotide identity to a strain detected in wild mice in New York City but less than 50% nucleotide identity to MuAstV, the strain commonly isolated from laboratory mice. In addition, MuAstV2 was found to share as much as 80.8% nucleotide similarity to an astrovirus strain isolated from urban brown rats (Rattus norvegicus) in Hong Kong.5,31 Antibodies to MuAstV2 were inadvertently detected in laboratory colony mice when a serologic immunoassay for mouse thymic virus prepared from a murine T-cell line tested positive. Further analysis showed that the mice were negative for mouse thymic virus and that the T-cell line was contaminated with a novel astrovirus strain similar to MuAstV2, resulting in the positive test. The observation that MuAstV2 did not appear to infect highly immunocompromised mice via natural exposure or experimental inoculation was highly unusual.31 This finding is distinct from other murine viruses, including MuAstV, given that infection of immunocompromised mice leads to persistent infection and chronic virus shedding.12,15,16,47We sought to further understand the biology of MuAstV2 by evaluating viral shedding kinetics and tissue tropism in immunocompetent mice and to further characterize the presumptive resistance to infection observed in highly immunocompromised mice. Temporal patterns of viral shedding were determined by serially measuring fecal viral RNA after oral inoculation. Tissue and cell tropism were characterized using in-situ hybridization (ISH) and immunohistochemistry during the course of infection. We hypothesized that MuAstV2 initially infects the gastrointestinal tract, as occurs with other astroviruses, but speculated that components of the immune system were required to support infection or replication or both. Furthermore, we sought to characterize the extraintestinal spread of MuAstV2.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号