Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Commun ; 14(1): 4206, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37452013

ABSTRACT

Hemophilia A is the most common X-linked bleeding disorder affecting more than half-a-million individuals worldwide. Persons with severe hemophilia A have coagulation FVIII levels <1% and experience spontaneous debilitating and life-threatening bleeds. Advances in hemophilia A therapeutics have significantly improved health outcomes, but development of FVIII inhibitory antibodies and breakthrough bleeds during therapy significantly increase patient morbidity and mortality. Here we use sheep fetuses at the human equivalent of 16-18 gestational weeks, and we show that prenatal transplantation of human placental cells (107-108/kg) bioengineered to produce an optimized FVIII protein, results in considerable elevation in plasma FVIII levels that persists for >3 years post-treatment. Cells engraft in major organs, and none of the recipients mount immune responses to either the cells or the FVIII they produce. Thus, these studies attest to the feasibility, immunologic advantage, and safety of treating hemophilia A prior to birth.


Subject(s)
Hemophilia A , Humans , Animals , Female , Pregnancy , Sheep , Hemophilia A/genetics , Factor VIII/genetics , Factor VIII/metabolism , Placenta/metabolism , Blood Coagulation , Fetus/metabolism
2.
Am J Physiol Renal Physiol ; 318(4): F861-F869, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32003597

ABSTRACT

Serum soluble Fas (sFas) levels are associated with erythropoietin (Epo) hyporesponsiveness in patients with chronic kidney disease (CKD). Whether sFas could predict the need for erythropoiesis-stimulating agent (ESA) usage and its influence in erythropoiesis remain unclear. We evaluated the relation between sFas and ESA therapy in patients with CKD with anemia and its effect on erythropoiesis in vitro. First, we performed a retrospective cohort study with 77 anemic patients with nondialysis CKD. We performed in vitro experiments to investigate whether sFas could interfere with the behavior of hematopoietic stem cells (HSCs). HSCs were isolated from umbilical cord blood and incubated with recombinant sFas protein in a dose-dependent manner. Serum sFas positively correlated with Epo levels (r = 0.30, P = 0.001) but negatively with hemoglobin (r = -0.55, P < 0.001) and glomerular filtration rate (r = -0.58, P < 0.001) in patients with CKD at baseline. Elevated sFas serum levels (4,316 ± 897 vs. 2,776 ± 749, P < 0.001) with lower estimated glomerular filtration rate (26.2 ± 10.1 vs. 33.5 ± 14.3, P = 0.01) and reduced hemoglobin concentration (11.1 ± 0.9 vs. 12.5 ± 1.2, P < 0.001) were identified in patients who required ESA therapy compared with patients with non-ESA. Afterward, we detected that the sFas level was slight correlated with a necessity of ESA therapy in patients with nondialysis CKD and anemia. In vitro assays demonstrated that the erythroid progenitor cell frequency negatively correlated with sFas concentration (r = -0.72, P < 0.001). There was decreased erythroid colony formation in vitro when CD34+ HSCs were incubated with a higher concentration of sFas protein (1.56 ± 0.29, 4.33 ± 0.53, P < 0.001). Our findings suggest that sFas is a potential predictor for ESA therapy in patients with nondialysis CKD and that elevated sFas could affect erythropoiesis in vitro.


Subject(s)
Anemia/blood , Erythropoiesis , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Renal Insufficiency, Chronic/complications , fas Receptor/blood , Adult , Aged , Anemia/diagnosis , Anemia/drug therapy , Anemia/etiology , Biomarkers/blood , Brazil , Cells, Cultured , Clinical Decision-Making , Databases, Factual , Erythropoiesis/drug effects , Erythropoietin/blood , Female , Hematinics/therapeutic use , Hematopoietic Stem Cells/drug effects , Hemoglobins/metabolism , Humans , Male , Middle Aged , Multipotent Stem Cells/drug effects , North Carolina , Patient Selection , Predictive Value of Tests , Recombinant Proteins/pharmacology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Retrospective Studies
3.
Int J Biol Sci ; 16(3): 435-446, 2020.
Article in English | MEDLINE | ID: mdl-32015680

ABSTRACT

Purpose: Drug-induced nephrotoxicity can occur in patients with pre-existing renal dysfunction or renal ischemia, potentially leading to chronic kidney disease (CKD) and end-stage renal disease (ESRD). Prompt treatment of CKD and the related side effects is critical in preventing progression to ESRD. The goal of this study was to demonstrate the therapeutic potential of urine-derived stem cells (USC) to treat chronic kidney disease-induced by nephrotoxic drugs and renal ischemia. Materials and methods: Human USC were collected, expanded and characterized by flow cytometry. A CKD model was induced by creating an ischemia-reperfusion injury and gentamicin administration. Twenty-eight adult immunodeficient rats were divided into three groups: PBS-treated group (n=9), USC-treated group (n=9), and sham group with age-matched control animals (n=10). Cell suspension of USC (5 x 106 / 100µl / kidney) or PBS was injected bilaterally into the renal parenchyma 9 weeks after CKD model creation. Renal function was evaluated by collection blood and urine samples to measure serum creatinine and glomerulus filtration rate. The kidneys were harvested 12 weeks after cell injection. Histologically, the extent of glomerulosclerosis and tubular atrophy, the amount of collagen deposition, interstitial fibrosis, inflammatory monocyte infiltration, and expression of transforming growth factor beta 1 (TGF-ß1), and superoxide dismutase 1 (SOD-1) were examined. Results: USC expressed renal parietal epithelial cells (CD24, CD29 and CD44). Renal function, measured by GFR and serum Cr in USC-treated group were significantly improved compared to PBS-treated animals (p<0.05). The degree of glomerular sclerosis and atrophic renal tubules, the amount of fibrosis, and monocyte infiltration significantly decreased in USC-treated group compared to the PBS group (p<0.05). The level of TGF-ß1 expression in renal tissues was also significantly lower in the PBS group, while the level of SOD-1 expression was significantly elevated in the USC group, compared to PBS group (p<0.05). Conclusions: The present study demonstrates the nephron-protective effect of USC on renal function via anti-inflammatory, anti-oxidative stress, and anti-fibrotic activity in a dual-injury CKD rat model. This provides an alternative treatment for CKD in certain clinical situations, such as instances where CKD is due to drug-induced nephrotoxicity and renal ischemia.


Subject(s)
Cell Differentiation/physiology , Renal Insufficiency, Chronic/therapy , Reperfusion Injury/therapy , Adipogenesis/physiology , Animals , Fibrosis/metabolism , Fibrosis/therapy , Humans , Ischemia/metabolism , Ischemia/therapy , Kidney/metabolism , Kidney/pathology , Male , Osteogenesis/physiology , Rats , Rats, Nude , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/metabolism
4.
Cardiovasc J Afr ; 31(4): 65-70, 2020.
Article in English | MEDLINE | ID: mdl-31591635

ABSTRACT

AIM: It has been postulated that gastro-oesophageal reflux disease (GORD) may trigger coronary ischaemia through viscerocardiac reflex vasoconstriction in subjects with ischaemic heart disease (IHD). Our aim was to estimate the prevalence of GORD in subjects with IHD who present with acute coronary syndrome (ACS) and to determine whether GORD may serve as a trigger for ischaemic events. METHODS: Twenty patients with isolated reflux oesophagitis and 39 with acute coronary syndrome (ACS with concomitant GORD) were studied. Twenty-two subjects comprising normal volunteers and those who were admitted for minor surgical trauma were used as normal controls. All subjects underwent oesophago-gastroduodenal endoscopy (EGD) and acid instillation with hydrochloric acid (0.1 M), as well as nuclear imaging (sestaMIBI) with technetium99. Ischaemia was detected by ST depression using ECG monitoring for one hour during and immediately after EGD. RESULTS: Of the 111 subjects with ACS, 39 (35.1%) had erosive GORD and comprised the study group. Subjects with ACS had more incidence of diabetes (p = 0.001), hypertension (p = 0.002), a history of smoking (p = 0.006) and elevated serum triglyceride levels (p = 0.008) compared to the GORD group. Risk-factor clustering in the form of the metabolic syndrome was more common in ACS subjects (44 vs 5%; p = 0.008). ST depression was documented in 8/39 (20.5%) patients in the ACS group and 5/20 (25%) in the GORD group (p = 0.958). Reversible perfusion defects on sestaMIBI scan were seen in 35.6% of the ACS subjects. CONCLUSIONS: Although GORD is common in subjects with ACS, we have not been able to show that GORD may serve as a trigger for ischaemia in these subjects.


Subject(s)
Acute Coronary Syndrome/epidemiology , Esophagitis, Peptic/epidemiology , Gastroesophageal Reflux/epidemiology , Acute Coronary Syndrome/diagnosis , Adult , Case-Control Studies , Electrocardiography , Endoscopy, Gastrointestinal , Esophagitis, Peptic/diagnosis , Female , Gastroesophageal Reflux/diagnosis , Humans , Male , Middle Aged , Prevalence , Radiopharmaceuticals/administration & dosage , Risk Assessment , Risk Factors , South Africa/epidemiology , Technetium Tc 99m Sestamibi/administration & dosage , Tomography, Emission-Computed, Single-Photon
5.
Small ; 15(43): e1902971, 2019 10.
Article in English | MEDLINE | ID: mdl-31464364

ABSTRACT

Human hematopoietic niches are complex specialized microenvironments that maintain and regulate hematopoietic stem and progenitor cells (HSPC). Thus far, most of the studies performed investigating alterations of HSPC-niche dynamic interactions are conducted in animal models. Herein, organ microengineering with microfluidics is combined to develop a human bone marrow (BM)-on-a-chip with an integrated recirculating perfusion system that consolidates a variety of important parameters such as 3D architecture, cell-cell/cell-matrix interactions, and circulation, allowing a better mimicry of in vivo conditions. The complex BM environment is deconvoluted to 4 major distinct, but integrated, tissue-engineered 3D niche constructs housed within a single, closed, recirculating microfluidic device system, and equipped with cell tracking technology. It is shown that this technology successfully enables the identification and quantification of preferential interactions-homing and retention-of circulating normal and malignant HSPC with distinct niches.


Subject(s)
Bone Marrow/metabolism , Cell Communication , Hematopoietic Stem Cells/pathology , Lab-On-A-Chip Devices , Stem Cell Niche , Antigens, CD34/metabolism , Biomarkers/metabolism , Cell Line, Tumor , Fluorescent Dyes/metabolism , Humans , Microtechnology
6.
Stem Cells Transl Med ; 8(9): 959-970, 2019 09.
Article in English | MEDLINE | ID: mdl-31144785

ABSTRACT

Renal disease is a worldwide health issue. Besides transplantation, current therapies revolve around dialysis, which only delays disease progression but cannot replace other renal functions, such as synthesizing erythropoietin. To address these limitations, cell-based approaches have been proposed to restore damaged kidneys as an alternative to current therapies. Recent studies have shown that stem cell-derived secretomes can enhance tissue regeneration. However, many growth factors undergo rapid degradation when they are injected into the body in a soluble form. Efficient delivery and controlled release of secreting factors at the sites of injury would improve the efficacy in tissue regeneration. Herein, we developed a gel-based delivery system for controlled delivery of trophic factors in the conditioned medium (CM) secreted from human placental stem cells (HPSCs) and evaluated the effect of trophic factors on renal regeneration. CM treatment significantly enhanced cell proliferation and survival in vitro. Platelet-rich plasma (PRP) was used as a delivery vehicle for CM. Analysis of the release kinetics demonstrated that CM delivery through the PRP gel resulted in a controlled release of the factors both in vitro and in vivo. In an acute kidney injury model in rats, functional and structural analysis showed that CM delivery using the PRP gel system into the injured kidney minimized renal tissue damage, leading to a more rapid functional recovery when compared with saline, CM, or vehicle only injection groups. These results suggest that controlled delivery of HPSC-derived trophic factors may provide efficient repair of renal tissue injury. Stem Cells Translational Medicine 2019;8:959&970.


Subject(s)
Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Kidney/drug effects , Animals , Apoptosis/drug effects , Cell Hypoxia , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Female , Gels/chemistry , Kidney/cytology , Kidney/pathology , Male , Placenta/cytology , Platelet-Rich Plasma/chemistry , Pregnancy , Rats , Rats, Nude , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Stem Cells/cytology , Stem Cells/metabolism
7.
Tissue Eng Part A ; 25(21-22): 1493-1503, 2019 11.
Article in English | MEDLINE | ID: mdl-30829146

ABSTRACT

Kidney disease is a major medical problem globally. Chronic kidney disease (CKD) is a progressive loss of kidney function. It causes accumulation of waste and fluid in the body, eventually resulting in kidney failure as well as damaging other organs. Although dialysis and kidney transplantation have been used as primary treatments for renal disease, dialysis does not restore full renal function, and there is a shortage of donor kidneys for transplantation. Recent advances in cell-based therapies have offered a means to augment and restore renal function. Various types of cells have been tested to evaluate their therapeutic effects on injured kidneys. Among various types of cells, amniotic fluid stem cells (AFSCs) share advantages of both embryonic and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects, which may allow their future therapeutic use as an "off-the-shelf" cell source. AFSC presents advantages of both conventional pluripotent and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects. This study demonstrates that administration of human-derived AFSC facilitates functional and structural improvement in a rat model of CKD, and suggests that cell therapy with AFSC has potential as a therapeutic strategy to recover renal function in patients with CKD. Impact Statement Patients with chronic kidney disease (CKD) have limited treatment options, and renal transplantation is the only definitive treatment method that restores kidney function. However, challenges associated with transplantation, including donor organ shortage, rejection, and life-long immunosuppression, remain a problem. Recently, stem cell-based therapies have been proposed as an alternative approach to augment and restore renal function. In this study, we used human-derived amniotic fluid stem cells (AFSCs) to treat CKD in a rat model and demonstrated that AFSC treatment facilitated positive effects in terms of improvements of renal function.


Subject(s)
Amniotic Fluid/cytology , Kidney Function Tests , Kidney/physiopathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Disease Models, Animal , Humans , Kidney/pathology , Male , Podocytes/ultrastructure , Rats, Nude
8.
Glob Adv Health Med ; 7: 2164956118777472, 2018.
Article in English | MEDLINE | ID: mdl-29796339

ABSTRACT

BACKGROUND: Interest in the use of extremely low-frequency (ELF) electromagnetic field (EMF) for the treatment of pain and inflammation is increasing due to the ability of this promising therapy to compete with pharmaceuticals without the adverse effects caused by drugs. However, there continues to be concerns regarding cytotoxic and genotoxic effects that may occur as a result of exposure to EMF. OBJECTIVE: To investigate this concern, we tested the effect of our known therapeutic 5 Hz, 0.4 milliTesla (mT) EMF on a human mesenchymal stromal cell (hMSC) line to determine whether ELF-EMF exposure would cause cytotoxic or genotoxic effects. METHODS: Treated samples along with controls were exposed to 5 Hz, 0.4 mT ELF-EMF for 20 min/day, 3×/week for 2 weeks and then assayed for cell viability, proliferation rates, and chromosome breaks. RESULTS: Cytogenetic analysis of the viability and proliferation rates along with analysis of morphological genome stability showed no cytotoxicity, and no chromosome breaks per karyotype analysis-therefore no genotoxicity. CONCLUSION: Exposure to an ELF-EMF of 5 Hz, 0.4 mT for 20 min/day, 3×/week for 2 weeks does not cause cytotoxic or genotoxic effects in hMSCs.

9.
PLoS One ; 11(10): e0164997, 2016.
Article in English | MEDLINE | ID: mdl-27776163

ABSTRACT

Chronic kidney disease (CKD) occurs when certain conditions cause the kidneys to gradually lose function. For patients with CKD, renal transplantation is the only treatment option that restores kidney function. In this study, we evaluated primary renal cells obtained from diseased kidneys to determine whether their normal phenotypic and functional characteristics are retained, and could be used for cell therapy. Primary renal cells isolated from both normal kidneys (NK) and diseased kidneys (CKD) showed similar phenotypic characteristics and growth kinetics. The expression levels of renal tubular cell markers, Aquaporin-1 and E-Cadherin, and podocyte-specific markers, WT-1 and Nephrin, were similar in both NK and CKD kidney derived cells. Using fluorescence- activated cell sorting (FACS), specific renal cell populations were identified and included proximal tubular cells (83.1% from NK and 80.3% from CKD kidneys); distal tubular cells (11.03% from NK and 10.9% from CKD kidneys); and podocytes (1.91% from NK and 1.78% from CKD kidneys). Ultra-structural analysis using scanning electron microscopy (SEM) revealed microvilli on the apical surface of cultured cells from NK and CKD samples. Moreover, transmission electron microscopy (TEM) analysis showed a similar organization of tight junctions, desmosomes, and other intracellular structures. The Na+ uptake characteristics of NK and CKD derived renal cells were also similar (24.4 mmol/L and 25 mmol/L, respectively) and no significant differences were observed in the protein uptake and transport characteristics of these two cell isolates. These results show that primary renal cells derived from diseased kidneys such as CKD have similar structural and functional characteristics to their counterparts from a normal healthy kidney (NK) when grown in vitro. This study suggests that cells derived from diseased kidney may be used as an autologous cell source for renal cell therapy, particularly in patients with CKD or end-stage renal disease (ESRD).


Subject(s)
Biomarkers/metabolism , Cell- and Tissue-Based Therapy/methods , Kidney/cytology , Renal Insufficiency, Chronic/therapy , Adolescent , Adult , Aged , Cell Separation , Female , Flow Cytometry , Humans , Kidney/metabolism , Kidney/ultrastructure , Male , Microscopy, Electron, Scanning , Middle Aged , Regenerative Medicine , Transplantation, Autologous
10.
Mol Cell Biol ; 32(21): 4350-62, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22927639

ABSTRACT

The function of MEX3C, the mammalian homolog of Caenorhabditis elegans RNA-binding protein muscle excess 3 (MEX-3), was unknown until our recent report that MEX3C is necessary for normal postnatal growth and enhances the expression of local bone Igf1 expression. Here we report the pivotal role of Mex3c in energy balance regulation. Mex3c mutation caused leanness in both heterozygous and homozygous transgenic mice, as well as a more beneficial blood glucose and lipid profile in homozygous transgenic mice, in both sexes. Although transgenic mice showed normal food intake and fecal lipid excretion, they had increased energy expenditure independent of physical activity. Mutant mice had normal body temperature, Ucp1 expression in brown adipose tissue, and muscle and liver fatty acid oxidation. Mex3c is expressed in neurons and is detectable in the arcuate nucleus, the ventromedial nucleus, and the dorsomedial nucleus of the hypothalamus. Mex3c was not detected in NPY or POMC neurons but was detected in leptin-responsive neurons in the ventromedial nucleus. Mex3c and Leptin double mutant mice were growth retarded and obese and had blood profiles similar to those of ob/ob mice but showed none of the steatosis observed in ob/ob mice. Our data show that Mex3c is involved in energy balance regulation.


Subject(s)
Adiposity/genetics , Energy Metabolism/genetics , Mutation , RNA-Binding Proteins/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Blood Glucose/analysis , Eating , Female , Ion Channels/biosynthesis , Leptin/deficiency , Leptin/genetics , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/biosynthesis , Neurons/metabolism , Obesity/genetics , Uncoupling Protein 1
11.
Biomaterials ; 33(29): 7261-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22795541

ABSTRACT

Pluripotent stem cells provide an unlimited cell source for cell therapy. However, residual pluripotent stem cells after differentiation can form tumors. Modifying stem cells with suicide constructs through integrating plasmid DNA and viral vectors has been attempted to specifically eliminate residual pluripotent stem cells after differentiation. However, integration of foreign DNA has the potential of insertional mutagenesis, position effects and silencing. Scaffold/matrix attachment region (S/MAR)-based plasmid DNA can be maintained extra-chromosomally, offering a safer alternative to integrating vectors for this purpose. Here, we report the design of an S/MAR-based suicide construct capable of episomal maintenance and specifically killing pluripotent stem cells but not differentiated cells in the presence of ganciclovir. Treating cells differentiated from episomal suicide construct-modified stem cells with ganciclovir reduces the tumor formation risk after cell transplantation. Tumors formed by such modified pluripotent stem cells could be inhibited by ganciclovir administration. This episomal suicide construct enables negative selection of residual pluripotent stem cells in vitro and control of tumors formed from residual pluripotent stem cells in vivo.


Subject(s)
Plasmids/chemistry , Pluripotent Stem Cells/cytology , Animals , CHO Cells , Cell Transplantation/methods , Cricetinae , DNA/chemistry , Female , Flow Cytometry/methods , Ganciclovir/administration & dosage , Ganciclovir/pharmacology , Gene Silencing , Genetic Engineering , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mice , Mice, SCID , Microscopy, Fluorescence/methods , Mutagenesis , Plasmids/metabolism , Stem Cells/cytology , Transgenes
12.
Free Radic Biol Med ; 52(11-12): 2223-33, 2012.
Article in English | MEDLINE | ID: mdl-22569411

ABSTRACT

Mitochondrial reactive oxygen species (ROS) have been implicated in spermatogenic damage, although direct in vivo evidence is lacking. We recently generated a mouse in which the inner mitochondrial membrane peptidase 2-like (Immp2l) gene is mutated. This Immp2l mutation impairs the processing of signal peptide sequences from mitochondrial cytochrome c1 and glycerol phosphate dehydrogenase 2. The mitochondria from mutant mice generate elevated levels of superoxide ion, which causes age-dependent spermatogenic damage. Here we confirm age-dependent spermatogenic damage in a new cohort of mutants, which started at the age of 10.5 months. Compared with age-matched controls, protein carbonyl content was normal in testes of 2- to 5-month-old mutants, but significantly elevated in testes of 13-month-old mutants, indicating elevated oxidative stress in the testes at the time of impaired spermatogenesis. Testicular expression of superoxide dismutases was not different between control and mutant mice, whereas that of catalase was increased in young and old mutants. The expression of cytosolic glutathione peroxidase 4 (phospholipid hydroperoxidase) in testes was significantly reduced in 13-month-old mutants, concomitant with impaired spermatogenesis. Apoptosis of all testicular populations was increased in mutant mice with spermatogenic damage. The mitochondrial DNA (mtDNA) mutation rate in germ cells of mutant mice with impaired spermatogenesis was unchanged, excluding a major role of mtDNA mutation in ROS-mediated spermatogenic damage. Our data show that increased mitochondrial ROS are one of the driving forces for spermatogenic impairment.


Subject(s)
Age Factors , Apoptosis , Endopeptidases/genetics , Mitochondrial Proteins/genetics , Oxidative Stress , Testis/metabolism , Animals , Apoptosis/genetics , Down-Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Male , Mice , Mice, Transgenic , Mutation/genetics , Oxidative Stress/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase , Protein Carbonylation/genetics , Spermatogenesis/genetics , Superoxides/metabolism , Testis/pathology
13.
Aging Cell ; 10(4): 584-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21332923

ABSTRACT

Mitochondrial reactive oxygen species (ROS) are proposed to play a central role in aging and age-associated disorders, although direct in vivo evidence is lacking. We recently generated a mouse mutant with mutated inner mitochondrial membrane peptidase 2-like (Immp2l) gene, which impairs the signal peptide sequence processing of mitochondrial proteins cytochrome c1 and glycerol phosphate dehydrogenase 2. The mitochondria from mutant mice generate elevated levels of superoxide ion and cause impaired fertility in both sexes. Here, we design experiments to examine the effects of excessive mitochondrial ROS generation on health span. We show that Immp2l mutation increases oxidative stress in multiple organs such as the brain and the kidney, although expression of superoxide dismutases in these tissues of the mutants is also increased. The mutants show multiple aging-associated phenotypes, including wasting, sarcopenia, loss of subcutaneous fat, kyphosis, and ataxia, with female mutants showing earlier onset and more severe age-associated disorders than male mutants. The loss of body weight and fat was unrelated to food intake. Adipose-derived stromal cells (ADSC) from mutant mice showed impaired proliferation capability, formed significantly less and smaller colonies in colony formation assays, although they retained adipogenic differentiation capability in vitro. This functional impairment was accompanied by increased levels of oxidative stress. Our data showed that mitochondrial ROS is the driving force of accelerated aging and suggested that ROS damage to adult stem cells could be one of the mechanisms for age-associated disorders.


Subject(s)
Adult Stem Cells/pathology , Aging, Premature/genetics , Endopeptidases/genetics , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Adult Stem Cells/metabolism , Aging, Premature/metabolism , Aging, Premature/pathology , Animals , Ataxia/genetics , Ataxia/metabolism , Endopeptidases/metabolism , Female , Kyphosis/genetics , Kyphosis/metabolism , Male , Mice , Mice, Mutant Strains , Mitochondria/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Mutation , Oxidative Stress , Reactive Oxygen Species/metabolism , Sarcopenia/genetics , Sarcopenia/metabolism , Sex Factors , Wasting Syndrome/genetics , Wasting Syndrome/metabolism
14.
J Virol Methods ; 172(1-2): 54-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21192980

ABSTRACT

The propagation of Taura syndrome virus (TSV) in primary hemocyte culture of Pacific white shrimp (Penaeus vannamei) was investigated. Purified TSV was inoculated into a 24 h old primary hemocyte culture and the development of cytopathic effects was monitored. The cell morphology started changing within 6 h post-inoculation; TSV-infected hemocytes started shrinking and granular structures began to form on the cell surface. There was a gradual loss of cell viability and, by 48 h post-inoculation, most cells detached from the bottom of the 96 well microplate. The propagation of TSV during the 48 h time course studied was measured by real-time RT-PCR. TSV copy number reached the highest level by 12 h post-inoculation and then started to decrease. Using an anti-TSV polyclonal antibody, the 55 kDa VP1 capsid protein was detected by Western blot analysis. The data suggest that shrimp primary hemocyte culture supports TSV replication and could be used as a tool for the study of host-virus interactions in TSV pathogenesis.


Subject(s)
Dicistroviridae/physiology , Penaeidae/virology , Virus Replication/physiology , Animals , Capsid Proteins/metabolism , Cell Survival , Cells, Cultured , Cytopathogenic Effect, Viral , Hemocytes/pathology , Hemocytes/virology , Reverse Transcriptase Polymerase Chain Reaction
15.
In Vitro Cell Dev Biol Anim ; 46(9): 801-10, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20835775

ABSTRACT

Improved methods of cell culture from eye stalk, hepatopancreas, muscle, ovary, and hemocytes of shrimp (Penaeus vannamei) were established using synthetic media and shrimp muscle extract (SME). For hemocytes and ovarian cell cultures, Grace's insect medium supplemented with 10% (v/v) fetal bovine serum and 10% SME (v/v) showed enhanced attachment and proliferation of the cells. The hemocyte and ovarian cell cultures could be maintained for 48 and 66 days, respectively, and have been sub-cultured four and six times, respectively. Both ovary and hemocyte cell cultures contained primarily epithelial-like cells. Cells derived from ovary tissue grew preferably between 26°C and 28°C with 5% CO(2). Although the temperature preference of hemocyte cells was the same as ovarian cells, CO(2) supplementation did not show any difference in the growth of hemocyte cells. When the shrimp were injected with lipopolysaccharide (8 µg/g of shrimp) and hemolymph was drawn 24 h post-injection, the in vitro multiplicity of hemocytes dramatically improved. The growth of eye stalk, hepatopancreas, and muscle-derived cells was much less compared to ovarian cells and hemocytes under the conditions described above. The optimal culture conditions for ovarian cells and hemocytes were also different from that for eye stalk, hepatopancreas, and muscle cell culture. The proliferation efficiencies of primary cultures of hepatopancreas, eyestalk, and muscle cells were about 30, 12, and <7 d, respectively. The improved culture conditions described here, particularly for hemocytes and ovary, will be very useful for in vitro studies involving viruses infecting shrimp and in shrimp genomic studies.


Subject(s)
Cell Culture Techniques/methods , Hemocytes/cytology , Hepatopancreas/cytology , Muscles/cytology , Ovary/cytology , Penaeidae/cytology , Posterior Eye Segment/cytology , Animals , Carbon Dioxide/chemistry , Female , Lipopolysaccharides , Specific Pathogen-Free Organisms , Temperature
16.
J Autoimmun ; 23(2): 151-60, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15324933

ABSTRACT

Little is known about the fate of autoreactive CD4 T cells in blood. Using a mouse model for spontaneous autoimmune diabetes we demonstrated that the status of the autoimmune process in pancreas could be pictured through the frequency and phenotype of autoreactive CD4 T cells in the blood. Early during the prediabetic stage, the frequency of these cells in blood decreased as a consequence of their recruitment in the pancreas. This was followed by an imbalance between CD4(+)CD25(+) and CD4(+)CD69(+) T cells in the pancreas that was mirrored in the phenotype of autoreactive T cells in the blood. Waves of activated CD4(+)CD69(+) T cells in blood preceded the disease onset suggesting that the autoimmune attack on pancreas is a discontinuous "hit-and-run" rather than a continuous process. Tracking autoreactive CD4 T cells in blood may help in identifying prediabetic humans and monitoring the disease progression during therapeutic interventions.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus/immunology , Pancreatic Diseases/immunology , Animals , Antigens, CD , Antigens, Differentiation, T-Lymphocyte , Blood Cells/immunology , Diabetes Complications/etiology , Diabetes Complications/immunology , Disease Models, Animal , Disease Progression , Immunophenotyping , Kinetics , Lectins, C-Type , Lymphocyte Count , Mice , Mice, Knockout , Mice, Transgenic , Pancreatic Diseases/etiology , Receptors, Interleukin-2 , T-Lymphocytes/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...