Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
iScience ; 26(7): 107122, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37416482

ABSTRACT

Gut dysbiosis has been associated with lupus pathogenesis, and fecal microbiota transfers (FMT) from lupus-prone mice shown to induce autoimmune activation into healthy mice. The immune cells of lupus patients exhibit an increased glucose metabolism and treatments with 2-deoxy-D-glucose (2DG), a glycolysis inhibitor, are therapeutic in lupus-prone mice. Here, we showed in two models of lupus with different etiologies that 2DG altered the composition of the fecal microbiome and associated metabolites. In both models, FMT from 2DG-treated mice protected lupus-prone mice of the same strain from the development of glomerulonephritis, reduced autoantibody production as well as the activation of CD4+ T cells and myeloid cells as compared to FMT from control mice. Thus, we demonstrated that the protective effect of glucose inhibition in lupus is transferable through the gut microbiota, directly linking alterations in immunometabolism to gut dysbiosis in the hosts.

2.
iScience ; 26(5): 106774, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37216123

ABSTRACT

The expansion of follicular helper T (Tfh) cells, which is tightly associated with the development of lupus, is reversed by the inhibition of either glycolysis or glutaminolysis in mice. Here we analyzed the gene expression and metabolome of Tfh cells and naive CD4+ T (Tn) cells in the B6.Sle1.Sle2.Sle3 (triple congenic, TC) mouse model of lupus and its congenic B6 control. Lupus genetic susceptibility in TC mice drives a gene expression signature starting in Tn cells and expanding in Tfh cells with enhanced signaling and effector programs. Metabolically, TC Tn and Tfh cells showed multiple defective mitochondrial functions. TC Tfh cells also showed specific anabolic programs including enhanced glutamate metabolism, malate-aspartate shuttle, and ammonia recycling, as well as altered dynamics of amino acid content and their transporters. Thus, our study has revealed specific metabolic programs that can be targeted to specifically limit the expansion of pathogenic Tfh cells in lupus.

3.
Biomolecules ; 12(10)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36291555

ABSTRACT

Human alpha 1 antitrypsin (hAAT) is a multifunctional protein that has been shown to have anti-inflammatory and cellular protective properties. While previous studies demonstrated the antiaging potential of hAAT, the mechanism(s) underlying the antiaging effect remain elusive. In this study, we performed a detailed analysis of transcriptomic data that indicated that NF-κB-targeted genes and NF-κB-regulated pathways were selectively inhibited by hAAT treatment. We further showed that the first detectable impact of hAAT treatment was the inhibition of the nuclear activity of NF-κB. Subsequently, hAAT treatment suppressed the mRNA levels of NF-κB-targeted genes, as well as NF-κB itself (P65 and P50), in human senescent cells. Using Drosophila models, we further examined the impact of hAAT on locomotor activity and endurance. Finally, using an adult-specific promotor, we demonstrated that overexpression of hAAT in the late stage of life significantly extended the lifespan of transgenic flies. These results extend the current understanding of the anti-inflammatory function of hAAT.


Subject(s)
Longevity , alpha 1-Antitrypsin , Animals , Humans , alpha 1-Antitrypsin/metabolism , Longevity/genetics , NF-kappa B/genetics , Drosophila/metabolism , RNA, Messenger
4.
Front Immunol ; 13: 914468, 2022.
Article in English | MEDLINE | ID: mdl-35860280

ABSTRACT

We report a novel model of lupus-associated cardiovascular pathology accelerated by the TLR7 agonist R848 in lupus-prone B6.Sle1.Sle2.Sle3 (TC) mice. R848-treated TC mice but not non-autoimmune C57BL/6 (B6) controls developed microvascular inflammation and myocytolysis with intracellular vacuolization. This histopathology was similar to antibody-mediated rejection after heart transplant, although it did not involve complement. The TC or B6 recipients of serum or splenocytes from R848-treated TC mice developed a reactive cardiomyocyte hypertrophy, which also presents spontaneously in old TC mice as well as in TC.Rag-/- mice that lack B and T cells. Each of these cardiovascular lesions correspond to abnormalities that have been reported in lupus patients. Lymphoid and non-lymphoid immune cells as well as soluble factors contribute to lupus-associated cardiovascular lesions in TC mice, which can now be dissected using this model with and without R848 treatment.


Subject(s)
Membrane Glycoproteins/metabolism , T-Lymphocytes , Toll-Like Receptor 7/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL
5.
J Immunol ; 208(5): 1066-1075, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35140132

ABSTRACT

BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Lymphoid Progenitor Cells/cytology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , T Follicular Helper Cells/immunology , Adoptive Transfer , Animals , Autoantibodies/immunology , B7-1 Antigen/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , Dendritic Cells/transplantation , Female , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/biosynthesis , Lymphocyte Activation/immunology , Lymphoid Progenitor Cells/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology
6.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34156979

ABSTRACT

Estrogen-related receptor γ (Esrrg) is a murine lupus susceptibility gene associated with T cell activation. Here, we report that Esrrg controls Tregs through mitochondria homeostasis. Esrrg deficiency impaired the maintenance and function of Tregs, leading to global T cell activation and autoimmunity in aged mice. Further, Esrrg-deficient Tregs presented an impaired differentiation into follicular Tregs that enhanced follicular helper T cells' responses. Mechanistically, Esrrg-deficient Tregs presented with dysregulated mitochondria with decreased oxygen consumption as well as ATP and NAD+ production. In addition, Esrrg-deficient Tregs exhibited decreased phosphatidylinositol and TGF-ß signaling pathways and increased mTOR complex 1 activation. We found that the expression of human ESRRG, which is high in Tregs, was lower in CD4+ T cells from patients with lupus than in healthy controls. Finally, knocking down ESRRG in Jurkat T cells decreased their metabolism. Together, our results reveal a critical role of Esrrg in the maintenance and metabolism of Tregs, which may provide a genetic link between lupus pathogenesis and mitochondrial dysfunction in T cells.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Mitochondria/pathology , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mice , Mitochondria/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
7.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32493841

ABSTRACT

Patients with systemic lupus erythematosus (SLE) present a high incidence of atherosclerosis, which contributes significantly to morbidity and mortality in this autoimmune disease. An impaired balance between regulatory (Treg) and follicular helper (Tfh) CD4+ T cells is shared by both diseases. However, whether there are common mechanisms of CD4+ T cell dysregulation between SLE and atherosclerosis remains unclear. Pre-B cell leukemia transcription factor 1 isoform d (Pbx1d) is a lupus susceptibility gene that regulates Tfh cell expansion and Treg cell homeostasis. Here, we investigated the role of T cells overexpressing Pbx1d in low-density lipoprotein receptor-deficient (Ldlr-/-) mice fed with a high-fat diet, an experimental model for atherosclerosis. Pbx1d-transgenic T cells exacerbated some phenotypes of atherosclerosis, which were associated with higher autoantibody production, increased Tfh cell frequency, and impaired Treg cell regulation, in Ldlr-/- mice as compared with control T cells. In addition, we showed that dyslipidemia and Pbx1d-transgenic expression independently impaired the differentiation and function of Treg cells in vitro, suggesting a gene/environment additive effect. Thus, our results suggest that the combination of Pbx1d expression in T cells and dyslipidemia exacerbates both atherosclerosis and autoimmunity, at least in part through a dysregulation of Treg cell homeostasis.


Subject(s)
Alleles , Atherosclerosis , Dyslipidemias , Gene Expression Regulation/immunology , Pre-B-Cell Leukemia Transcription Factor 1/immunology , T-Lymphocytes, Regulatory , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Dyslipidemias/genetics , Dyslipidemias/immunology , Dyslipidemias/pathology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Pre-B-Cell Leukemia Transcription Factor 1/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
8.
Immunohorizons ; 4(6): 319-331, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32540987

ABSTRACT

CTLA4Ig, a reagent that inhibits CD28 signaling, has shown therapeutic efficacy in mouse models of lupus nephritis (LN) when combined with several other biologics or standard of care drugs. Unfortunately, clinical trials treating LN patients with CTLA4Ig (abatacept) have not met endpoints. Metformin, a drug used to control hyperglycemia that inhibits mitochondrial metabolism, lowered the effective dose of glucocorticoids and prevented major flares when added on to the standard of care treatment of lupus patients with low disease activity. Metformin combined with inhibition of glycolysis by 2-deoxyglucose showed therapeutic efficacy in multiple mouse models of LN. Because CD28 signaling triggers glucose metabolism in T cells, we hypothesized that combining CTLA4Ig treatment with metformin would have the same effect. In this study, we showed that the combination of metformin and CTLA4Ig decreased the development of LN in (NZB × NZW)F1 mice treated at the early stage of disease. This preventive effect was associated with a decreased expansion of CD4+ T cell effector subsets. However, contrary to the combination with 2-deoxyglucose, metformin combined with CTLA4Ig did not alter autoantibody production, suggesting different mechanisms of symptom mitigation. Overall, this study shows therapeutic efficacy of the combination of metformin and CTLA4Ig, two drugs with established safety records, in a preclinical mouse model of LN.


Subject(s)
Abatacept/pharmacology , Immunosuppressive Agents/pharmacology , Lupus Nephritis/immunology , Lupus Nephritis/prevention & control , Metformin/pharmacology , Animals , Antigens, CD , Autoantibodies/blood , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Disease Models, Animal , Drug Therapy, Combination , Female , Kidney/drug effects , Kidney/pathology , Lupus Nephritis/drug therapy , Lymphocyte Activation , Mice , Mice, Inbred NZB , T-Lymphocyte Subsets/immunology
9.
J Clin Med ; 8(9)2019 Aug 29.
Article in English | MEDLINE | ID: mdl-31470606

ABSTRACT

Diffuse alveolar hemorrhage (DAH) is a fatal complication in patients with lupus. DAH can be induced in B6 mice by an intraperitoneal injection of pristane. Since human alpha-1-antitrypsin (hAAT) is an anti-inflammatory and immuno-regulatory protein, we investigated the protective effect of hAAT against pristane-induced DAH in B6 mice and hAAT transgenic (hAAT-Tg) mice. We first showed that hAAT Tg expression lowers TNF-α production in B cells, as well as CD4+ T cells in untreated mice. Conversely, the frequency of regulatory CD4+CD25+ and CD4+CD25-IL-10+ cells was significantly higher in hAAT-Tg than in B6 mice. This confirmed the anti-inflammatory effect of hAAT that was observed even at steady state. One week after a pristane injection, the frequency of peritoneal Ly6Chi inflammatory monocytes and neutrophils in hAAT-Tg mice was significantly lower than that in B6 mice. Importantly, pristane-induced DAH was completely prevented in hAAT-Tg mice and this was associated with a modulation of anti- to pro-inflammatory myeloid cell ratio/balance. We also showed that treatment with hAAT decreased the severity of DAH in B6 mice. These results showed for the first time that hAAT has a therapeutic potential for the treatment of DAH.

10.
Methods Mol Biol ; 1826: 143-155, 2018.
Article in English | MEDLINE | ID: mdl-30194599

ABSTRACT

Alpha-1-antitrypsin (AAT) is a circulating protein, a serpin, with multiple protective functions. Beside the well-known proteinase inhibitory function, which protects the lungs from chronic obstructive pulmonary disease (COPD), many studies have shown that AAT inhibits pro-inflammatory cytokine gene expression and functions. These anti-inflammatory and immune-regulatory properties have led to studies testing the therapeutic effect of AAT in autoimmune disease models. Initially, a study using recombinant adeno-associated viral (rAAV) vector showed that AAT gene therapy prevented type 1 diabetes (T1D) development in a nonobese diabetic (NOD) mouse model. Consequently, several studies confirmed that AAT therapy prevented and reversed T1D. AAT therapy has also been tested and has demonstrated protective effects in a collagen-induced arthritis model and a systemic lupus erythematosus (SLE) mouse model. This chapter describes methods that evaluate AAT functions in autoimmune mouse models.


Subject(s)
Arthritis, Experimental , Dependovirus , Diabetes Mellitus, Type 1 , Genetic Therapy , Lupus Erythematosus, Systemic , alpha 1-Antitrypsin , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Inbred NOD , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/therapy , Transduction, Genetic , alpha 1-Antitrypsin/biosynthesis , alpha 1-Antitrypsin/genetics
11.
Aging Cell ; 17(1)2018 02.
Article in English | MEDLINE | ID: mdl-29045001

ABSTRACT

Inflammaging plays an important role in most age-related diseases. However, the mechanism of inflammaging is largely unknown, and therapeutic control of inflammaging is challenging. Human alpha-1 antitrypsin (hAAT) has immune-regulatory, anti-inflammatory, and cytoprotective properties as demonstrated in several disease models including type 1 diabetes, arthritis, lupus, osteoporosis, and stroke. To test the potential anti-inflammaging effect of hAAT, we generated transgenic Drosophila lines expressing hAAT. Surprisingly, the lifespan of hAAT-expressing lines was significantly longer than that of genetically matched controls. To understand the mechanism underlying the anti-aging effect of hAAT, we monitored the expression of aging-associated genes and found that aging-induced expressions of Relish (NF-ĸB orthologue) and Diptericin were significantly lower in hAAT lines than in control lines. RNA-seq analysis revealed that innate immunity genes regulated by NF-kB were significantly and specifically inhibited in hAAT transgenic Drosophila lines. To confirm this anti-inflammaging effect in human cells, we treated X-ray-induced senescence cells with hAAT and showed that hAAT treatment significantly decreased the expression and maturation of IL-6 and IL-8, two major factors of senescence-associated secretory phenotype. Consistent with results from Drosophila,RNA-seq analysis also showed that hAAT treatment significantly inhibited inflammation related genes and pathways. Together, our results demonstrated that hAAT significantly inhibited inflammaging in both Drosophila and human cell models. As hAAT is a FDA-approved drug with a confirmed safety profile, this novel therapeutic potential may make hAAT a promising candidate to combat aging and aging-related diseases.


Subject(s)
Aging/physiology , Inflammation/drug therapy , Osteoporosis/drug therapy , alpha 1-Antitrypsin/pharmacology , Animals , Drosophila , Genetic Therapy/methods , Longevity/drug effects
12.
Mol Med ; 23: 57-69, 2017 05.
Article in English | MEDLINE | ID: mdl-28332697

ABSTRACT

Osteoporosis is a global public health problem affecting more than 200 million people worldwide. We previously showed that treatment with alpha-1 antitrypsin (AAT), a multifunctional protein with anti-inflammatory properties, mitigated bone loss in an ovariectomized mouse model. However, the underlying mechanisms of the protective effect of AAT on bone tissue are largely unknown. In this study, we investigated the effect of AAT on osteoclast formation and function in vitro. Our results showed that AAT dose-dependently inhibited the formation of RANKL (receptor activator of nuclear factor κB ligand) induced osteoclasts derived from mouse bone marrow macrophages/monocyte (BMM) lineage cells and the murine macrophage cell line, RAW 264.7 cells. In order to elucidate the possible mechanisms underlying this inhibition, we tested the effect of AAT on the gene expression of cell surface molecules, transcription factors, and cytokines associated with osteoclast formation. We showed that AAT inhibited M-CSF (macrophage colony-stimulating factor) induced cell surface RANK expression in osteoclast precursor cells. In addition, AAT inhibited RANKL-induced TNF-α production, cell surface CD9 expression, and dendritic cell-specific transmembrane protein (DC-STAMP) gene expression. Importantly, AAT treatment significantly inhibited osteoclast-associated mineral resorption. Together, these results uncovered new mechanisms for the protective effects of AAT and strongly support the notion that AAT has therapeutic potential for the treatment of osteoporosis.


Subject(s)
Osteoclasts/drug effects , alpha 1-Antitrypsin/pharmacology , Animals , Bone Marrow Cells/cytology , Cytokines/metabolism , Macrophage Colony-Stimulating Factor , Male , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , Osteoporosis/drug therapy , RANK Ligand , RAW 264.7 Cells
13.
Hum Gene Ther ; 28(2): 179-189, 2017 02.
Article in English | MEDLINE | ID: mdl-27802778

ABSTRACT

Osteoporosis is a common health problem severely affecting the quality of life of many people, especially women. Current treatment options for osteoporosis are limited due to their association with several side-effects and moderate efficacy. Therefore, novel therapies for osteoporosis are needed. This study tested the feasibility of adipose tissue-derived mesenchymal stem cell (ATMSC)-based human alpha-1 antitrypsin (hAAT, SERPINA1) gene therapy for the prevention of bone loss in an ovariectomized (OVX) mouse model. Eight-week-old female C57BL6 mice underwent ovariectomy and were treated with hAAT (protein therapy), ATMSC (stem-cell therapy), ATMSC + hAAT (combination of ATMSC and hAAT therapy), and ATMSCs infected with lentiviral vectors expressing hAAT (ATMSC-based hAAT gene therapy). The study showed that lenti-hAAT vector-infected ATMSCs (ATMSC-LV-hAAT) produced high levels of hAAT. Transplantation of these cells significantly decreased OVX-induced serum levels of interleukin 6 and interleukin 1 beta, and receptor activator of nuclear factor kappa B gene expression levels in bone tissue. Immunohistological analysis revealed that transplanted cells migrated to the bone tissue and secreted hAAT. Importantly, bone microstructure analysis by microcomputerized tomography showed that this treatment significantly protected against OVX-induced bone loss. The results suggest a novel strategy for the treatment of osteoporosis in humans.


Subject(s)
Adipose Tissue/cytology , Genetic Therapy , Genetic Vectors/administration & dosage , Lentivirus/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Osteoporosis/therapy , alpha 1-Antitrypsin/genetics , Animals , Bone Density , Combined Modality Therapy , Disease Models, Animal , Mice , Osteoporosis/etiology , Ovariectomy/adverse effects
14.
Hum Gene Ther ; 27(9): 679-86, 2016 09.
Article in English | MEDLINE | ID: mdl-27158796

ABSTRACT

Osteoporosis is a major healthcare burden affecting mostly postmenopausal women characterized by compromised bone strength and increased risk of fragility fracture. Although pathogenesis of this disease is complex, elevated proinflammatory cytokine production is clearly involved in bone loss at menopause. Therefore, anti-inflammatory strategies hold a great potential for the prevention of postmenopausal osteoporosis. In this study, we investigated the effect of gene therapy of recombinant adeno-associated virus (rAAV)-mediated human alpha-1 antitrypsin (hAAT), a multifunctional protein that has anti-inflammatory property, on bone loss in an ovariectomy-induced osteoporosis mouse model. Adult ovariectomized (OVX) mice were intraperitoneally (i.p.) injected with hAAT (protein therapy), rAAV8-CB-hAAT (gene therapy), or phosphate buffer saline (PBS). Age-matched and sham-operated animals were used as controls. Eight weeks after the treatment, animals were sacrificed and bone-related biomarkers and vertebral bone structure were evaluated. Results showed that hAAT gene therapy significantly decreased serum IL-6 level and receptor activator of NF-κB (RANK) gene expression in bone. Importantly, hAAT gene therapy increased bone volume/total volume and decreased structure model index (SMI) compared to PBS injection in OVX mice. These results demonstrate that hAAT gene therapy by rAAV vector efficiently mitigates bone loss possibly through inhibition of proinflammatory cytokine IL-6 and RANK gene expression. Considering the safety profile of hAAT and rAAV vector in humans, our results provide a new alternative for the treatment of osteoporosis.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Osteoporosis/prevention & control , Ovariectomy/adverse effects , alpha 1-Antitrypsin/genetics , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Osteoporosis/etiology
15.
PLoS One ; 11(5): e0156583, 2016.
Article in English | MEDLINE | ID: mdl-27232337

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disorder with a worldwide distribution and considerable mortality and morbidity. Although the pathogenesis of this disease remains elusive, over-reactive dendritic cells (DCs) play a critical role in the disease development. It has been shown that human alpha-1 antitrypsin (hAAT) has protective effects in type 1 diabetes and rheumatoid arthritis mouse models. In the present study, we tested the effect of AAT on DC differentiation and functions, as well as its protective effect in a lupus-prone mouse model. We showed that hAAT treatment significantly inhibited LPS (TLR4 agonist) and CpG (TLR9 agonist) -induced bone-marrow (BM)-derived conventional and plasmacytoid DC (cDC and pDC) activation and reduced the production of inflammatory cytokines including IFN-I, TNF-α and IL-1ß. In MRL/lpr mice, hAAT treatment significantly reduced BM-derived DC differentiation, serum autoantibody levels, and importantly attenuated renal pathology. Our results for the first time demonstrate that hAAT inhibits DC activation and function, and it also attenuates autoimmunity and renal damage in the MRL/lpr lupus model. These results imply that hAAT has a therapeutic potential for the treatment of SLE in humans.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/drug effects , Lupus Erythematosus, Systemic/immunology , alpha 1-Antitrypsin/pharmacology , Animals , Autoantibodies/biosynthesis , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cytokines/biosynthesis , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation/drug effects , Humans , Lupus Erythematosus, Systemic/metabolism , Male , Mice
16.
PLoS One ; 10(9): e0138184, 2015.
Article in English | MEDLINE | ID: mdl-26372641

ABSTRACT

OBJECTIVE: Adipose tissue derived stem cells (ADSCs) transplantation has recently gained widespread enthusiasm, particularly in the perspective to use them as potential alternative cell sources for hepatocytes in cell based therapy, mainly because of their capability of hepatogenic differentiation in vitro and in vivo. But some challenges remain to be addressed, including whether ADSCs can be provided effectively to the target organ and whether subsequent proliferation of transplanted cells can be achieved. To date, intrasplenic injection is the conventional method to deliver ADSCs into the liver; however, a number of donor cells retained in the spleen has been reported. In this study, our objective is to evaluate a novel route to transplant ADSCs specifically to the liver. We aimed to test the feasibility of in situ transplantation of ADSCs by injecting bioencapsulated ADSCs into the liver in mouse model. METHODS: The ADSCs isolated from human alpha 1 antitrypsin (M-hAAT) transgenic mice were used to allow delivered ADSCs be readily identified in the liver of recipient mice, and alginate was selected as a cell carrier. We first evaluated whether alginate microspheres are implantable into the liver tissue by injection and whether ADSCs could migrate from alginate microspheres (study one). Once proven, we then examined the in vivo fate of ADSCs loaded microspheres in the liver. Specifically, we evaluated whether transplanted, undifferentiated ASDCs could be induced by the local microenvironment toward hepatogenic differentiation and the distribution of surviving ADSCs in major tissue organs (study two). RESULTS: Our results indicated ADSCs loaded alginate microspheres were implantable into the liver. Both degraded and residual alginate microspheres were observed in the liver up to three weeks. The viable ADSCs were detectable surrounding degraded and residual alginate microspheres in the liver and other major organs such as bone marrow and the lungs. Importantly, transplanted ADSCs underwent hepatogenic differentiation to become cells expressing albumin in the liver. These findings improve our understanding of the interplay between ADSCs (donor cells), alginate (biomaterial), and local microenvironment in a hepatectomized mouse model, and might improve the strategy of in situ transplantation of ADSCs in treating liver diseases.


Subject(s)
Adipose Tissue/cytology , Alginates/chemistry , Liver/cytology , Stem Cell Transplantation/methods , Stem Cells/chemistry , Stem Cells/cytology , Animals , Capsules , Cell Differentiation , Feasibility Studies , Glucuronic Acid/chemistry , Hepatocytes/cytology , Hexuronic Acids/chemistry , Humans , Injections , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microspheres , alpha 1-Antitrypsin/genetics
17.
J Stroke Cerebrovasc Dis ; 23(5): e355-63, 2014.
Article in English | MEDLINE | ID: mdl-24582784

ABSTRACT

Our objective is to develop a new therapy for the treatment of stroke. Currently, the only effective therapy for acute ischemic stroke is the thrombolytic agent recombinant tissue plasminogen activator. α1-Antitrypsin (AAT), a serine proteinase inhibitor with potent anti-inflammatory, anti-apoptotic, antimicrobial, and cytoprotective activities, could be beneficial in stroke. The goal of this study is to test whether AAT can improve ischemic stroke outcome in an established rat model. Middle cerebral artery occlusion was induced in male rats via intracranial (i.c.) microinjection of endothelin-1. Five to 10 minutes after stroke induction, rats received either i.c. or intravenous delivery of human AAT. Cylinder and vibrissae tests were used to evaluate sensorimotor function before and 72 hours after middle cerebral artery occlusion. Infarct volumes were examined via either 2,3,5-triphenyltetrazolium chloride assay or magnetic resonance imaging 72 hours after middle cerebral artery occlusion. Despite equivalent initial strokes, at 72 hours, the infarct volumes of the human AAT treatment groups (local and systemic injection) were statistically significantly reduced by 83% and 63% (P < .0001 and P < .05, respectively) compared with control rats. Human AAT significantly limited sensory motor system deficits. Human AAT could be a potential novel therapeutic drug for the protection against neurodegeneration after ischemic stroke, but more studies are needed to investigate the protective mechanisms and efficacy in other animal models.


Subject(s)
Brain/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , alpha 1-Antitrypsin/pharmacology , Animals , Brain/blood supply , Brain/pathology , Brain/physiopathology , Cytoprotection , Disease Models, Animal , Endothelin-1 , Humans , Infarction, Middle Cerebral Artery/chemically induced , Infarction, Middle Cerebral Artery/diagnosis , Infarction, Middle Cerebral Artery/physiopathology , Injections, Intravenous , Magnetic Resonance Imaging , Male , Microinjections , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Rats, Sprague-Dawley , Sensory Thresholds/drug effects , Time Factors , alpha 1-Antitrypsin/administration & dosage
18.
Hum Gene Ther Methods ; 25(1): 72-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24191859

ABSTRACT

Induced pluripotent stem (iPS) cells have great potential for personalized regenerative medicine. Although several different methods for generating iPS cells have been reported, improvement of safety and efficiency is imperative. In this study, we tested the feasibility of using a triple tyrosine mutant AAV2 (Y444+500+730F) vector, designated AAV2.3m, to generate iPS cells. We developed a polycistronic rAAV2.3m vector expressing three reprogramming factors, Klf4, Oct4, and Sox2, and then used this vector to infect mouse adipose-derived mesenchymal stem cells (AT-MSCs) to induce the generation of iPS cells. We demonstrated that (1) the triple tyrosine mutant AAV2 vector is able to reprogram mouse adult adipose tissue-derived stem cells into the pluripotent state. Those rAAV2.3m-derived iPS (rAAV2.3m-iPS) cells express endogenous pluripotency-associated genes including Oct4, Sox2, and SSEA-1, and form teratomas containing multiple tissues in vivo; (2) c-myc, an oncogene, is dispensable in rAAV2.3m-mediated cellular reprogramming; and (3) transgene expression is undetectable after reprogramming, whereas vector DNA is detectable, indicating that transgenes are silenced. These results indicated the rAAV vector may have some advantages in generating iPS cells.


Subject(s)
Adipose Tissue/cytology , Cellular Reprogramming , Dependovirus/genetics , Genetic Vectors/metabolism , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Genetic Vectors/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Teratoma/pathology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...