Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Genet Metab ; 96(3): 133-44, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19131261

ABSTRACT

The MRL mouse is an inbred laboratory strain that was derived by selective breeding in 1960 from the rapidly growing LG/J (Large) strain. MRL mice grow to nearly twice the size of other commonly used mouse strains, display uncommonly robust healing and regeneration properties, and express later onset autoimmune traits similar to Systemic Lupus Erythematosis. The regeneration trait (heal) in the MRL mouse maps to 14-20 quantitative trait loci and the autoimmune traits map to 5-8 loci. In this paper we report the metabolic and biochemical features that characterize the adult MRL mouse and distinguish it from C57BL/6 control animals. We found that adult MRL mice have retained a number of features of embryonic metabolism that are normally lost during development in other strains. These include an emphasis on aerobic glycolytic energy metabolism, increased glutamate oxidation, and a reduced capacity for fatty acid oxidation. MRL tissues, including the heart, liver, and regenerating ear hole margins, showed considerable mitochondrial genetic and physiologic reserve, decreased mitochondrial transmembrane potential (DeltaPsi(m)), decreased reactive oxygen species (ROS), and decreased oxidative phosphorylation, yet increased mitochondrial DNA and protein content. The discovery of embryonic metabolic features led us to look for cells that express markers of embryonic stem cells. We found that the adult MRL mouse has retained populations of cells that express the stem cell markers Nanog, Islet-1, and Sox2. These are present in the heart at baseline and highly induced after myocardial injury. The retention of embryonic features of metabolism in adulthood is rare in mammals. The MRL mouse provides a unique experimental window into the relationship between metabolism, stem cell biology, and regeneration.


Subject(s)
Mice, Inbred MRL lpr/embryology , Mice, Inbred MRL lpr/metabolism , Animals , Embryonic Stem Cells/metabolism , Fatty Acids/metabolism , Female , Glutathione/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr/genetics , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
2.
Clin Immunol ; 92(3): 300-10, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479535

ABSTRACT

Wound healing is a complex process that involves inflammation, apoptosis, growth, and tissue remodeling. The autoimmune-prone inbred mouse strain MRL/+ manifests accelerated and extensive healing to ear punch wounds, suggesting a link between immune defects and wound healing. Prior studies with lupus-prone mice have shown that hematopoietic cells of lupus-prone strains can transfer disease to otherwise non-autoimmune-prone recipients. In this study we performed reciprocal bone marrow transfers between MRL and the control strain B10.BR and found that radioresistant MRL/+ host cells, rather than hematopoietic cells, are required for the healing response. We have also made the novel observations that, compared to normal controls, MRL/+ hematopoietic cells overproduce TGF-beta1 and manifest impaired inflammatory responses to lipopolysaccharide challenge. These features suggest that the aberrant wound healing phenotype of MRL mice is independent of their propensity to develop autoimmunity.


Subject(s)
Mice, Inbred MRL lpr/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/physiology , Aging/genetics , Aging/physiology , Animals , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Genotype , Hematopoietic Stem Cells/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Neutrophils/cytology , Pneumonia/physiopathology , Transplantation Chimera , Wound Healing/genetics
3.
Kidney Int ; 53(6): 1631-41, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9607194

ABSTRACT

We report that the beta-chemokine RANTES, a chemoattractant for macrophages and T cells, is up-regulated in the MRL-Fas(1pr) kidney prior to injury, but not normal kidneys (MRL-++, C3H-++) and increases with progressive injury. Furthermore, we establish an association between RANTES expression in the kidney and renal damage using a gene transfer approach. Tubular epithelial cells genetically modified to secrete RANTES infused under the renal capsule incites interstitial nephritis in MRL-Fas(1pr), but not MRL-++ or C3H-++ mice. RANTES recruits predominantly macrophages (M phi) and CD4+ and CD8+ T cells. In contrast, gene transfer of CSF-1, another molecule up-regulated simultaneously with RANTES in MRL-Fas(1pr) kidneys, promotes the influx of M phi, CD4+ T cells and the unique double-negative (DN) T cells (CD4-, CD8-), which are prominent in diseased MRL-Fas(1pr) kidneys. Thus, RANTES and CSF-1 recruit distinct T cell populations into the MRL-Fas(1pr) kidney. In addition, delivery of RANTES and CSF-1 into the kidney of MRL-Fas(1pr) mice causes an additive increase in pathology. We suggest that the complementary recruitment of T cell populations by RANTES (CD4, CD8) and CSF-1 (CD4, DN) promotes autoimmune nephritis in MRL-Fas(1pr) mice.


Subject(s)
Autoimmune Diseases/genetics , Chemokine CCL5/genetics , Gene Transfer Techniques , Kidney Diseases/genetics , Mice, Inbred MRL lpr/genetics , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , CD4 Antigens/analysis , CD8 Antigens/analysis , Chemokine CCL5/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/pathology , Mice , Mice, Inbred C3H , Mice, Inbred MRL lpr/metabolism , RNA, Messenger/metabolism , T-Lymphocytes/immunology
4.
Kidney Int ; 52(4): 934-41, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9328932

ABSTRACT

MRL-Faslpr mice are an appealing strain to understand the importance of cytokines in the pathogenesis of autoimmune renal destruction, since injury is rapid and predictable. Colony stimulating factor 1 (CSF-1) and tumor necrosis factor alpha (TNF-alpha) are detected in the kidney and circulation prior to renal injury and continue to increase with progressive renal damage in MRL-Faslpr, Fas deficient mice, but not the congenic Fas intact MRL-(++) strain. Delivery of CSF-1, but not TNF-alpha, into the kidney via gene transfer incites local renal injury. By comparison, dual gene transfer of CSF-1 and TNF-alpha incites autoimmune renal injury that is far more severe than CSF-1 alone. The purpose of this study was to establish whether CSF-1 and TNF-alpha incites autoimmune renal injury that is far more severe than CSF-1 alone. The purpose of this study was to establish whether CSF-1 and TNF-alpha expression in the kidney of MRL-Faslpr mice induced by a circulating stimulant resulted from a primary defect in the kidney. Therefore, we transplanted (Tx) a MRL-(++) kidney without CSF-1, TNF-alpha and renal injury into an MRL-Faslpr recipient after removing nephritic kidney expressing CSF-1 and TNF-alpha. The Tx kidneys were examined after 2, 4, 5, 6, and 12 weeks. CSF-1 and TNF-alpha were rapidly induced in the MRL-(++) Tx kidney. CSF-1 and TNF-alpha were evident by two weeks and continually increased for 12 weeks post-transplantation. Within several weeks, the rapid expansion of M phi and T cells and induction of glomerulonephritis and interstitial nephritis in the MRL-(++) Tx kidney was similar to the age-matched native MRL-Faslpr kidney. In conclusion, we have constructed an experimental transplantation system that can explore whether cytokine expression in the kidney induced by a circulating stimulant is a result of a primary defect in the kidney. Using this approach, we established that the MRL-Faslpr kidney is not defective, but rather a circulating stimulant in the MRL-Faslpr mouse can induce CSF-1, TNF-alpha and renal injury in a normal MRL-(++) kidney. Thus, we exclude an intrinsic defect in the MRL-Faslpr kidney as the pathogenic mechanism responsible for tissue damage. We suggest purging the circulation of molecules that induce CSF-1 and TNF-alpha as a therapeutic strategy for autoimmune renal injury.


Subject(s)
Autoimmune Diseases/metabolism , Kidney/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mice, Inbred MRL lpr/metabolism , Nephritis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Autoimmune Diseases/blood , Glomerulonephritis/etiology , Kidney Transplantation , Mice , Nephritis/blood , Nephritis, Interstitial/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...