Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Transplantation ; 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39228019

ABSTRACT

BACKGROUND: Severe primary graft dysfunction (PGD) after lung transplantation (LTx) is a significant risk factor for the development of bronchiolitis obliterans syndrome (BOS). Recent data from our group demonstrated that small extracellular vesicles (sEVs) isolated from the plasma of LTx recipients with BOS have reduced levels of tumor suppressor gene liver kinase B1 (LKB1) and promote epithelial-to-mesenchymal transition (EMT) and fibrosis. Here, we hypothesized that early inflammatory responses associated with severe PGD (PGD2/3) can downregulate LKB1 levels in sEVs, predisposing to the development of chronic lung allograft dysfunction (CLAD). METHODS: sEVs were isolated from the plasma of human participants by Exosome Isolation Kit followed by 0.20-µm filtration and characterized by NanoSight and immunoblotting analysis. Lung self-antigens (K alpha 1 tubulin, Collagen V), LKB1, nuclear factor kappa B, and EMT markers in sEVs were compared by densitometry analysis between PGD2/3 and no-PGD participants. Neutrophil-derived factors and hypoxia/reperfusion effects on LKB1 levels and EMT were analyzed in vitro using quantitative real-time polymerase chain reaction and Western blotting. RESULTS: LKB1 was significantly downregulated in PGD2/3 sEVs compared with no-PGD sEVs. Within PGD2/3 participants, lower post-LTx LKB1 was associated with CLAD development. Hypoxia/reperfusion downregulates LKB1 and is associated with markers of EMT in vitro. Finally, lower LKB1 levels in PGD2/3 are associated with increased markers of EMT. CONCLUSIONS: Our results suggest that in post-LTx recipients with PGD2/3, downregulation of LKB1 protein levels in sEVs is associated with increased EMT markers and may result in the development of CLAD. Our results also suggest that ischemia/reperfusion injury during LTx may promote CLAD through the early downregulation of LKB1.

2.
Cell Rep Med ; 4(3): 100945, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36787736

ABSTRACT

Accumulation of senescent cells contributes to age-related diseases including idiopathic pulmonary fibrosis (IPF). Insulin-like growth factor binding proteins (IGFBPs) regulate many biological processes; however, the functional contributions of IGFBP2 in lung fibrosis remain largely unclear. Here, we report that intranasal delivery of recombinant IGFBP2 protects aged mice from weight loss and demonstrated antifibrotic effects after bleomycin lung injury. Notably, aged human-Igfbp2 transgenic mice reveal reduced senescence and senescent-associated secretory phenotype factors in alveolar epithelial type 2 (AEC2) cells and they ameliorated bleomycin-induced lung fibrosis. Finally, we demonstrate that IGFBP2 expression is significantly suppressed in AEC2 cells isolated from fibrotic lung regions of patients with IPF and/or pulmonary hypertension compared with patients with hypersensitivity pneumonitis and/or chronic obstructive pulmonary disease. Altogether, our study provides insights into how IGFBP2 regulates AEC2-cell-specific senescence and that restoring IGFBP2 levels in fibrotic lungs can prove effective for patients with IPF.


Subject(s)
Alveolar Epithelial Cells , Idiopathic Pulmonary Fibrosis , Aged , Animals , Humans , Mice , Alveolar Epithelial Cells/metabolism , Bleomycin/adverse effects , Bleomycin/metabolism , Cellular Senescence/genetics , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Mice, Transgenic
3.
Cell Immunol ; 386: 104690, 2023 04.
Article in English | MEDLINE | ID: mdl-36812767

ABSTRACT

BACKGROUND: We recently demonstrated decreased tumor suppressor gene liver kinase B1 (LKB1) level in lung transplant recipients diagnosed with bronchiolitis obliterans syndrome. STE20-related adaptor alpha (STRADα) functions as a pseudokinase that binds and regulates LKB1 activity. METHODS: A murine model of chronic lung allograft rejection in which a single lung from a B6D2F1 mouse was orthotopically transplanted into a DBA/2J mouse was employed. We examined the effect of LKB1 knockdown using CRISPR-CAS9 in vitro culture system. RESULTS: Significant downregulation of LKB1 and STRADα expression was found in donor lung compared to recipient lung. STRADα knockdown significantly inhibited LKB1, pAMPK expression but induced phosphorylated mammalian target of rapamycin (mTOR), fibronectin, and Collagen-I, expression in BEAS-2B cells. LKB1 overexpression decreased fibronectin, Collagen-I, and phosphorylated mTOR expression in A549 cells. CONCLUSIONS: We demonstrated that downregulation of LKB1-STRADα pathway accompanied with increased fibrosis, results in development of chronic rejection following murine lung transplantation.


Subject(s)
Fibronectins , Lung Transplantation , Animals , Mice , Fibronectins/genetics , Fibronectins/metabolism , Down-Regulation , Mice, Inbred DBA , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Lung/metabolism , Biomarkers , Genes, Tumor Suppressor , Allografts , Collagen/genetics , Collagen/metabolism , Mammals/genetics , Mammals/metabolism
4.
Transpl Immunol ; 75: 101702, 2022 12.
Article in English | MEDLINE | ID: mdl-36038048

ABSTRACT

OBJECTIVE: Antibodies against donor human leukocyte antigen are a risk factor for chronic immune injury (CII) following renal transplantation; however, it is often not detectable. The main goal of this study is to gain new insights into the kinetics of exosome release and content in sensitized vs non-sensitized recipients. Towards this, we investigated the role for circulating exosomes with allo and self-antigens as well as immunoregulatory molecules in the development of CII and acute rejection. METHODS: Using murine kidney allograft rejection models, we investigated the role of exosomes on immune responses leading to allo- and auto-immunity to self-antigens resulting in rejection. Exosomes were analyzed for kidney self-antigens (Collagen-IV, fibronectin, angiotensin II receptor type 1), and immune-regulatory molecules (PD-L1, CD73) using western blot. Antibodies to donor MHC in serum samples were detected by immunofluorescence, self-antigens by enzyme-linked immunosorbent assay and kidney tissue infiltrating cells were determined by immunohistochemistry. RESULTS: BALB/c; H2d to C57BL/6; H2b renal transplantation (BALB/c), resulted in tubulitis and cellular infiltration by day 14, suggestive of acute inflammation, that was self-limiting with functioning graft. This contributed to CII on post-transplant day >100, which was preceded by induction of exosomes with donor and self-antigens leading to antibodies and immune-regulatory molecules. The absence of acute rejection in this allogenic transplant model is likely due to the induction of splenic and, graft-infiltrating CD4 + FoxP3+ T regulatory cells. In contrast, prior sensitization by skin graft followed by kidney transplantation induced antibodies to MHC and self-antigens leading to acute rejection. CONCLUSION: We demonstrate a pivotal role for induction of exosomes with immune-regulatory molecules, allo- and auto-immunity to self-antigens leading to chronic immune injury following murine kidney transplantation.


Subject(s)
Exosomes , Kidney Transplantation , Humans , Mice , Animals , Autoantigens , Graft Rejection , HLA Antigens , Mice, Inbred BALB C , Histocompatibility Antigens
5.
Front Immunol ; 13: 861583, 2022.
Article in English | MEDLINE | ID: mdl-35572510

ABSTRACT

Transplantation is a treatment option for patients diagnosed with end-stage organ diseases; however, long-term graft survival is affected by rejection of the transplanted organ by immune and nonimmune responses. Several studies have demonstrated that both acute and chronic rejection can occur after transplantation of kidney, heart, and lungs. A strong correlation has been reported between de novo synthesis of donor-specific antibodies (HLA-DSAs) and development of both acute and chronic rejection; however, some transplant recipients with chronic rejection do not have detectable HLA-DSAs. Studies of sera from such patients demonstrate that immune responses to tissue-associated antigens (TaAgs) may also play an important role in the development of chronic rejection, either alone or in combination with HLA-DSAs. The synergistic effect between HLA-DSAs and antibodies to TaAgs is being established, but the underlying mechanism is yet to be defined. We hypothesize that HLA-DSAs damage the transplanted donor organ resulting in stress and leading to the release of extracellular vesicles, which contribute to chronic rejection. These vesicles express both donor human leukocyte antigen (HLA) and non-HLA TaAgs, which can activate antigen-presenting cells and lead to immune responses and development of antibodies to both donor HLA and non-HLA tissue-associated Ags. Extracellular vesicles (EVs) are released by cells under many circumstances due to both physiological and pathological conditions. Primarily employing clinical specimens obtained from human lung transplant recipients undergoing acute or chronic rejection, our group has demonstrated that circulating extracellular vesicles display both mismatched donor HLA molecules and lung-associated Ags (collagen-V and K-alpha 1 tubulin). This review focuses on recent studies demonstrating an important role of antibodies to tissue-associated Ags in the rejection of transplanted organs, particularly chronic rejection. We will also discuss the important role of extracellular vesicles released from transplanted organs in cross-talk between alloimmunity and autoimmunity to tissue-associated Ags after solid organ transplantation.


Subject(s)
Extracellular Vesicles , Organ Transplantation , Antibodies , Autoantigens , Autoimmunity , Graft Rejection , HLA Antigens , Histocompatibility Antigens Class I , Humans
6.
Am J Transplant ; 22(3): 843-852, 2022 03.
Article in English | MEDLINE | ID: mdl-34859569

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been implicated to play a role in chronic lung allograft dysfunction (CLAD). Liver kinase B1 (LKB1), a tumor suppressor gene, can regulate EMT. However, its role in CLAD development following lung transplantation remains unknown. Using qRT-PCR, biopsies from lung transplant recipients with bronchiolitis obliterans syndrome (BOS) demonstrated significant downregulation of LKB1 (p = .0001), compared to stable biopsies. To determine the role of LKB1 in EMT development, we analyzed EMT in human bronchial epithelial cell line BEAS-2B. Knockdown of LKB1 by siRNA significantly dysregulated mesenchymal markers expression in BEAS-2B cells. Following incubation of human primary bronchial epithelial cell or BEAS-2B cells with exosomes isolated from BOS or stable lung transplant recipients, LKB1 expression was inhibited when incubated with BOS-exosome. Incubation with BOS-exosomes also decreased LKB1 expression and induced EMT markers in air-liquid interface culture method. Our results provide novel evidence that exosomes released from transplanted lungs undergoing chronic rejection are associated with inactivated tumor suppressor gene LKB1 and this loss induces EMT leading to the pathogenesis of CLAD following human lung transplantation.


Subject(s)
Bronchiolitis Obliterans , Graft vs Host Disease , Lung Transplantation , Allografts , Biomarkers , Bronchiolitis Obliterans/etiology , Epithelial-Mesenchymal Transition , Genes, Tumor Suppressor , Humans , Liver , Lung , Lung Transplantation/adverse effects
7.
Transpl Int ; 33(1): 41-49, 2020 01.
Article in English | MEDLINE | ID: mdl-31393646

ABSTRACT

Chronic lung allograft dysfunction (CLAD) comprises both bronchiolitis obliterans syndrome and restrictive allograft syndrome as subtypes. After lung transplantation, CLAD remains a major limitation for long-term survival, and lung transplant recipients therefore have poorer outcomes compared with recipients of other solid organ transplants. Although the number of lung transplants continues to increase globally, the field demands detailed understanding of immunoregulatory mechanisms and more effective individualized therapies to combat CLAD. Emerging evidence suggests that CLAD is multifactorial and involves a complex, delicate interplay of multiple factors, including perioperative donor characteristics, inflammation induced immediately following transplant, post-transplant infection and interplay between allo- and autoimmunity directed to donor antigens. Recently, identification of stress-induced exosome release from the transplanted organ has emerged as an underlying mechanism in the development of chronic rejection and promises to prompt novel strategies for future therapeutic interventions. In this review, we will discuss recent studies and ongoing research into the mechanisms for the development of CLAD, with emphasis on immune responses to lung-associated self-antigens-that is, autoimmunity.


Subject(s)
Autoantibodies , Bronchiolitis Obliterans , Lung Transplantation , Autoimmunity , Bronchiolitis Obliterans/etiology , Graft Rejection , Humans , Lung/physiopathology , Lung Transplantation/adverse effects
8.
Nat Commun ; 10(1): 5705, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836710

ABSTRACT

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Subject(s)
Capillaries/growth & development , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Kidney Glomerulus/blood supply , Animals , Capillaries/cytology , Capillaries/metabolism , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Kidney Glomerulus/growth & development , Kidney Glomerulus/metabolism , Male , Mice , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
9.
Crit Rev Immunol ; 39(2): 123-134, 2019.
Article in English | MEDLINE | ID: mdl-31679252

ABSTRACT

Exosomes, nanovesicles shown to regulate physiological processes in vivo, have been implicated in pathological conditions including cancer, autoimmune disease, infectious disease, neurodegenerative disease, and allograft rejection. Studies of lung transplant recipients with primary graft dysfunction, respiratory viral infection, and (acute) rejection have demonstrated circulating exosomes containing donor-mismatched human leukocyte antigen and lung-associated self-antigens, K-alpha 1 tubulin and collagen V, indicating that exosomes are originating from the transplanted organ. These circulating exosomes likely play a role in activating immune responses that lead to increased risk of chronic lung allograft dysfunction, as exosomes efficiently present their antigens to the immune system by all known pathways of antigen recognition (i.e., direct, indirect, and semidirect pathways). Here, we discuss exosome biogenesis, describe their contents, and address the mechanism of exosome-mediated activation of immune responses that lead to allograft rejection, especially after lung transplantation.


Subject(s)
Autoantigens/immunology , Collagen Type V/immunology , Exosomes/immunology , Graft Rejection/immunology , Lung Transplantation , Tubulin/immunology , Allografts/immunology , Animals , Blood Circulation , Exosomes/metabolism , HLA Antigens/immunology , Humans , Respiratory Tract Infections/immunology , Transplantation, Homologous , Virus Diseases/immunology
10.
JCI Insight ; 3(11)2018 06 07.
Article in English | MEDLINE | ID: mdl-29875323

ABSTRACT

Sepsis causes acute kidney injury (AKI) in critically ill patients, although the pathophysiology remains unclear. The receptor-interacting protein kinase-3 (RIPK3), a cardinal regulator of necroptosis, has recently been implicated in the pathogenesis of human disease. In mice subjected to polymicrobial sepsis, we demonstrate that RIPK3 promotes sepsis-induced AKI. Utilizing genetic deletion and biochemical approaches in vitro and in vivo, we identify a potentially novel pathway by which RIPK3 aggravates kidney tubular injury independently of the classical mixed lineage kinase domain-like protein-dependent (MLKL-dependent) necroptosis pathway. In kidney tubular epithelial cells, we show that RIPK3 promotes oxidative stress and mitochondrial dysfunction involving upregulation of NADPH oxidase-4 (NOX4) and inhibition of mitochondrial complex I and -III, and that RIPK3 and NOX4 are critical for kidney tubular injury in vivo. Furthermore, we demonstrate that RIPK3 is required for increased mitochondrial translocation of NOX4 in response to proinflammatory stimuli, by a mechanism involving protein-protein interactions. Finally, we observed elevated urinary and plasma RIPK3 levels in human patients with sepsis-induced AKI, representing potential markers of this condition. In conclusion, we identify a pathway by which RIPK3 promotes kidney tubular injury via mitochondrial dysfunction, independently of MLKL, which may represent a promising therapeutic target in sepsis-induced AKI.


Subject(s)
Acute Kidney Injury/pathology , Kidney Tubules/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Shock, Septic/complications , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Acute Kidney Injury/urine , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Biomarkers/blood , Biomarkers/urine , Cell Line , Epithelial Cells/cytology , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Female , Humans , Kidney Tubules/cytology , Kidney Tubules/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Middle Aged , Mitochondria/pathology , Mitochondria/ultrastructure , NADPH Oxidase 4/metabolism , Necrosis/pathology , Oxidative Stress , Primary Cell Culture , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/blood , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/urine , Shock, Septic/blood , Shock, Septic/urine , Up-Regulation , Young Adult
11.
Am J Physiol Renal Physiol ; 311(5): F877-F889, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27440777

ABSTRACT

Growth delay is common in children with chronic kidney disease (CKD), often associated with poor quality of life. The role of anemia in uremic growth delay is poorly understood. Here we describe an induction of uremic growth retardation by a 0.2% adenine diet in wild-type (WT) and hepcidin gene (Hamp) knockout (KO) mice, compared with their respective littermates fed a regular diet. Experiments were started at weaning (3 wk). After 8 wk, blood was collected and mice were euthanized. Adenine-fed WT mice developed CKD (blood urea nitrogen 82.8 ± 11.6 mg/dl and creatinine 0.57 ± 0.07 mg/dl) and were 2.1 cm shorter compared with WT controls. WT adenine-fed mice were anemic and had low serum iron, elevated Hamp, and elevated IL6 and TNF-α. WT adenine-fed mice had advanced mineral bone disease (serum phosphorus 16.9 ± 3.1 mg/dl and FGF23 204.0 ± 115.0 ng/ml) with loss of cortical and trabecular bone volume seen on microcomputed tomography. Hamp disruption rescued the anemia phenotype resulting in improved growth rate in mice with CKD, thus providing direct experimental evidence of the relationship between Hamp pathway and growth impairment in CKD. Hamp disruption ameliorated CKD-induced growth hormone-insulin-like growth factor 1 axis derangements and growth plate alterations. Disruption of Hamp did not mitigate the development of uremia, inflammation, and mineral and bone disease in this model. Taken together, these results indicate that an adenine diet can be successfully used to study growth in mice with CKD. Hepcidin appears to be related to pathways of growth retardation in CKD suggesting that investigation of hepcidin-lowering therapies in juvenile CKD is warranted.


Subject(s)
Anemia/metabolism , Growth Disorders/metabolism , Hepcidins/metabolism , Renal Insufficiency, Chronic/metabolism , Adenine , Anemia/diagnostic imaging , Anemia/genetics , Animals , Disease Models, Animal , Femur/diagnostic imaging , Fibroblast Growth Factor-23 , Growth Disorders/chemically induced , Growth Disorders/genetics , Growth Plate/diagnostic imaging , Hepcidins/genetics , Mice , Mice, Knockout , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/diagnostic imaging , Renal Insufficiency, Chronic/genetics , X-Ray Microtomography
12.
Am J Respir Cell Mol Biol ; 55(5): 722-735, 2016 11.
Article in English | MEDLINE | ID: mdl-27374190

ABSTRACT

Administration of supplemental oxygen remains a critical clinical intervention for survival of preterm infants with respiratory failure. However, prolonged exposure to hyperoxia can augment pulmonary damage, resulting in developmental lung diseases embodied as hyperoxia-induced acute lung injury and bronchopulmonary dysplasia (BPD). We sought to investigate the role of autophagy in hyperoxia-induced apoptotic cell death in developing lungs. We identified increased autophagy signaling in hyperoxia-exposed mouse lung epithelial-12 cells, freshly isolated fetal type II alveolar epithelial cells, lungs of newborn wild-type mice, and human newborns with respiratory distress syndrome and evolving and established BPD. We found that hyperoxia exposure induces autophagy in a Trp53-dependent manner in mouse lung epithelial-12 cells and in neonatal mouse lungs. Using pharmacological inhibitors and gene silencing techniques, we found that the activation of autophagy, upon hyperoxia exposure, demonstrated a protective role with an antiapoptotic response. Specifically, inhibiting regulatory-associated protein of mechanistic target of rapamycin (RPTOR) in hyperoxia settings, as evidenced by wild-type mice treated with torin2 or mice administered (Rptor) silencing RNA via intranasal delivery or Rptor+/-, limited lung injury by increased autophagy, decreased apoptosis, improved lung architecture, and increased survival. Furthermore, we identified increased protein expression of phospho-beclin1, light chain-3-II and lysosomal-associated membrane protein 1, suggesting altered autophagic flux in the lungs of human neonates with established BPD. Collectively, our study unveils a novel demonstration of enhancing autophagy and antiapoptotic effects, specifically through the inhibition of RPTOR as a potentially useful therapeutic target for the treatment of hyperoxia-induced acute lung injury and BPD in developing lungs.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/pathology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Autophagy , Hyperoxia/complications , Hyperoxia/pathology , Acute Lung Injury/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Autophagy/drug effects , Bronchopulmonary Dysplasia/complications , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Cell Line , Female , Humans , Hyperoxia/metabolism , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/pathology , Infant, Newborn , Lung/metabolism , Lung/pathology , Mice , Microtubule-Associated Proteins/metabolism , Naphthyridines/pharmacology , Phenotype , Regulatory-Associated Protein of mTOR , Time Factors , Tumor Suppressor Protein p53/metabolism
13.
Am J Physiol Renal Physiol ; 310(7): F596-F606, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26739888

ABSTRACT

Transforming growth factor-ß (TGF-ß) is generally considered as a central mediator of fibrotic diseases. Indeed, much focus has been placed on inhibiting TGF-ß and its downstream targets as ideal therapeutic strategies. However, pharmacological blockade of TGF-ß has not yet translated into successful therapy for humans, which may be due to pleiotropic effects of TGF-ß signaling. Equally, TGF-ß signaling as a protective response in kidney injury has been relatively underexplored. An emerging body of evidence from experimental kidney disease models indicates multifunctionality of TGF-ß capable of inducing profibrotic and protective effects. This review discusses recent advances highlighting the diverse roles of TGF-ß in promoting not only renal fibrosis but also protective responses of TGF-ß signaling. We review, in particular, growing evidence that supports protective effects of TGF-ß by mechanisms which include inhibiting inflammation and induction of autophagy. Additional detailed studies are required to fully understand the diverse mechanisms of TGF-ß actions in renal fibrosis and inflammation that will likely direct toward effective antifibrotic therapies.


Subject(s)
Kidney/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Kidney/pathology , Smad Proteins/metabolism
14.
Respir Res ; 16: 4, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25591994

ABSTRACT

BACKGROUND: Earlier studies have reported that transforming growth factor beta 1(TGFß1) is a critical mediator of hyperoxia-induced acute lung injury (HALI) in developing lungs, leading to impaired alveolarization and a pulmonary phenotype of bronchopulmonary dysplasia (BPD). However, the mechanisms responsible for the TGFß1-induced inflammatory signals that lead to cell death and abnormal alveolarization are poorly understood. We hypothesized that TGFß1 signaling via TGFßR2 is necessary for the pathogenesis of the BPD pulmonary phenotype resulting from HALI. METHODS: We utilized lung epithelial cell-specific TGFß1 overexpressing transgenic and TGFßR2 null mutant mice to evaluate the effects on neonatal mortality as well as pulmonary inflammation and apoptosis in developing lungs. Lung morphometry was performed to determine the impaired alveolarization and multicolor flow cytometry studies were performed to detect inflammatory macrophages and monocytes in lungs. Apoptotic cell death was measured with TUNEL assay, immunohistochemistry and western blotting and protein expression of angiogenic mediators were also analyzed. RESULTS: Our data reveals that increased TGFß1 expression in newborn mice lungs leads to increased mortality, macrophage and immature monocyte infiltration, apoptotic cell death specifically in Type II alveolar epithelial cells (AECs), impaired alveolarization, and dysregulated angiogenic molecular markers. CONCLUSIONS: Our study has demonstrated the potential role of inhibition of TGFß1 signaling via TGFßR2 for improved survival, reduced inflammation and apoptosis that may provide insights for the development of potential therapeutic strategies targeted against HALI and BPD.


Subject(s)
Acute Lung Injury/metabolism , Apoptosis , Lung/metabolism , Pneumonia/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/biosynthesis , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Animals, Newborn , Disease Models, Animal , Genotype , Humans , Hyperoxia/complications , Lung/pathology , Lung/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Pneumonia/genetics , Pneumonia/pathology , Pneumonia/physiopathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Time Factors , Transforming Growth Factor beta1/genetics , Up-Regulation
15.
Redox Biol ; 4: 208-14, 2015.
Article in English | MEDLINE | ID: mdl-25613291

ABSTRACT

Both acute kidney injury (AKI) and chronic kidney disease (CKD) that lead to diminished kidney function are interdependent risk factors for increased mortality. If untreated over time, end stage renal disease (ESRD) is an inevitable outcome. Acute and chronic kidney diseases occur partly due to imbalance between the molecular mechanisms that govern oxidative stress, inflammation, autophagy and cell death. Oxidative stress refers to the cumulative effects of highly reactive oxidizing molecules that cause cellular damage. Autophagy removes damaged organelles, protein aggregates and pathogens by recruiting these substrates into double membrane vesicles called autophagosomes which subsequently fuse with lysosomes. Mounting evidence suggests that both oxidative stress and autophagy are significantly involved in kidney health and disease. However, very little is known about the signaling processes that link them. This review is focused on understanding the role of oxidative stress and autophagy in kidney diseases. In this review, we also discuss the potential relationships between oxidative stress and autophagy that may enable the development of better therapeutic intervention to halt the progression of kidney disease and promote its repair and resolution.


Subject(s)
Acute Kidney Injury/metabolism , Autophagy , Kidney/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Gene Expression Regulation , Humans , Inflammation , Kidney/drug effects , Kidney/pathology , Lysosomes/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Phagosomes/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Signal Transduction
16.
Zebrafish ; 12(1): 33-47, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25517940

ABSTRACT

The Johns Hopkins Clinical Compound Library (JHCCL), a collection of Food and Drug Administration (FDA)-approved small molecules (1400), was screened in silico for identification of novel ß2AR blockers and tested for hematopoietic stem cell (HSC) expansion and radioprotection in zebrafish embryos. Docking studies, followed by the capacity to hasten erythropoiesis, identified todralazine (Binding energy, -8.4 kcal/mol) as a potential HSC-modulating agent. Todralazine (5 µM) significantly increased erythropoiesis in caudal hematopoietic tissue (CHT) in wild-type and anemic zebrafish embryos (2.33- and 1.44-folds, respectively) when compared with untreated and anemic control groups. Todralazine (5 µM) treatment also led to an increased number of erythroid progenitors, as revealed from the increased expression of erythroid progenitor-specific genes in the CHT region. Consistent with these effects, zebrafish embryos, Tg(cmyb:gfp), treated with 5 µM todralazine from 24 to 36 hours post fertilization (hpf) showed increased (approximately two-folds) number of HSCs at the aorta-gonad-mesonephros region (AGM). Similarly, expression of HSC marker genes, runx1 (3.3-folds), and cMyb (1.41-folds) also increased in case of todralazine-treated embryos, further supporting its HSC expansion potential. Metoprolol, a known beta blocker, also induced HSC expansion (1.36- and 1.48-fold increase in runx1 and cMyb, respectively). Todralazine (5 µM) when added 30 min before 20 Gy gamma radiation, protected zebrafish from radiation-induced organ toxicity, apoptosis, and improved survival (80% survival advantage over 6 days). The 2-deoxyribose degradation test further suggested hydroxyl (OH) radical scavenging potential of todralazine, and the same is recapitulated in vivo. These results suggest that todralazine is a potential HSC expanding agent, which might be acting along with important functions, such as antioxidant and free radical scavenging, in manifesting radioprotection.


Subject(s)
Adrenergic beta-2 Receptor Antagonists/pharmacology , Hematopoietic Stem Cells/drug effects , Radiation, Ionizing , Todralazine/pharmacology , Zebrafish/metabolism , Animals , Embryo, Nonmammalian/drug effects
17.
PLoS One ; 8(4): e60560, 2013.
Article in English | MEDLINE | ID: mdl-23637753

ABSTRACT

BACKGROUND: The role and mechanism of action of MIF in hyperoxia-induced acute lung injury (HALI) in the newborn lung are not known. We hypothesized that MIF is a critical regulatory molecule in HALI in the developing lung. METHODOLOGY: We studied newborn wild type (WT), MIF knockout (MIFKO), and MIF lung transgenic (MIFTG) mice in room air and hyperoxia exposure for 7 postnatal (PN) days. Lung morphometry was performed and mRNA and protein expression of vascular mediators were analyzed. RESULTS: MIF mRNA and protein expression were significantly increased in WT lungs at PN7 of hyperoxia exposure. The pattern of expression of Angiopoietin 2 protein (in MIFKO>WT>MIFTG) was similar to the mortality pattern (MIFKO>WT>MIFTG) in hyperoxia at PN7. In room air, MIFKO and MIFTG had modest but significant increases in chord length, compared to WT. This was associated with decreased expression of Angiopoietin 1 and Tie 2 proteins in the MIFKO and MIFTG, as compared to the WT control lungs in room air. However, on hyperoxia exposure, while the chord length was increased from their respective room air controls, there were no differences between the 3 genotypes. CONCLUSION: These data point to the potential roles of Angiopoietins 1, 2 and their receptor Tie2 in the MIF-regulated response in room air and upon hyperoxia exposure in the neonatal lung.


Subject(s)
Hyperoxia/complications , Lung Injury/etiology , Lung Injury/metabolism , Lung/growth & development , Lung/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Angiopoietins/genetics , Animals , Bronchoalveolar Lavage , Cell Count , Gene Expression Regulation , Gene Knockout Techniques , Interleukin-6/metabolism , Lung/pathology , Lung Injury/genetics , Lung Injury/pathology , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Transgenic , Phenotype , Receptor, TIE-2/genetics , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Front Physiol ; 4: 384, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24421769

ABSTRACT

During mild stressful conditions, cells activate a multitude of mechanisms in an attempt to repair or re-establish homeostasis. One such mechanism is autophagic degradation of mitochondria or mitophagy to dispose damaged mitochondria. However, if stress persists beyond recovery then dysfunctional mitochondria can ignite cell death. This review article summarizes recent studies highlighting the molecular pathways that facilitate mitochondria to alter its morphological dynamics, coordinate stress responses, initiate mitophagy and activate cell death in relevance to pulmonary pathologies. Thorough understanding of how these signaling mechanisms get disrupted may aid in designing new mitochondria-based therapies to combat lung diseases.

19.
J Cell Sci ; 125(Pt 7): 1693-705, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22328518

ABSTRACT

Maintenance of tissue boundaries is crucial for control of metastasis. We describe a new signalling pathway in which epithelial cell disruption can be minimised and thereby restricts epithelial-mesenchymal transgressions. This involves the release of insulin-like growth factor (IGF)-binding protein 5 (IGFBP5) from apoptotic cells, which increases the adhesion of epithelial cells on mesenchymal but not epithelial extracellular matrix (ECM), and involves the direct interaction of IGFBP5 and α2ß1 integrins. IGFBP5 also induced cell adhesion to vitronectin in the absence of αVß3 integrin, the vitronectin receptor, again through an α2ß1-integrin-dependent action, suggesting that IGFBP5 can induce spreading on matrices, even in the absence of the integrins normally used in this process. Using IGFBP5 mutants we demonstrate that the effect is IGF-independent but requires the heparin-binding domain in the C-terminus of IGFBP5. A truncated mutant containing only the C-terminal of IGFBP5 also induced adhesion. Adhesion induced by IGFBP5 was dependent on Cdc42 and resulted in activation of integrin-linked kinase (ILK) and Akt. Consistent with these changes, IGFBP5 facilitated prolonged cell survival in nutrient-poor conditions and decreased phosphorylation of the stress-activated kinase p38 MAPK (MAPK14). Whereas IGFBP5 enhanced adhesion, it inhibited cell migration, although this was not evident using the truncated C-terminal mutant, suggesting that effects of IGFBP5 on adhesion and migration involve different mechanisms. We anticipate that these responses to IGFBP5 would reduce the metastatic potential of cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Insulin-Like Growth Factor Binding Protein 5/metabolism , Cell Adhesion , Cell Survival , Female , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , MCF-7 Cells
20.
Int J Dev Biol ; 55(7-9): 781-9, 2011.
Article in English | MEDLINE | ID: mdl-22161834

ABSTRACT

Mammary gland development is dependent upon insulin-like growth factors (IGFs) as survival factors. The actions of the IGFs are modulated by a family of IGF-binding proteins (IGFBP1-6). Expression of the IGFBPs is both time-dependent and cell-specific during both the developmental phases and the involution of the mammary gland. Although studied extensively in vitro, understanding the roles of IGFBPs in vivo has been difficult, largely due to the fact that IGFBP knock-out mice have no dramatic phenotypes. This review examines the evidence from in vitro studies and the attempts to examine in vivo actions utilising models with IGFBP deficiency or over-expression. In vitro studies demonstrate that IGFBPs can act by inhibition of the survival effects of IGFs, as well as by enhancing the effects of IGFs. Because the IGFBPs are found associated with the extracellular matrix, a role for IGFBPs as a reservoir of IGFs or, alternatively as a potential barrier to IGFs, thereby restricting their entry into particular tissues or cellular compartments was postulated. We also provide evidence with respect to the IGF-independent actions of the IGFBPs which include receptors, nuclear localization, and interaction with the extracellular matrix and cell surface proteins including integrins. We believe that recent findings place some of the IGFBPs in a larger family of extracellular proteins, the secreted cysteine-rich protein (CCN) family, which have similar structural domains (involved in binding to IGFs, extracellular matrix and integrins) and are heavily implicated in tissue re-modeling and morphogenesis.


Subject(s)
Breast/growth & development , Insulin-Like Growth Factor Binding Proteins/physiology , Mammary Glands, Animal/growth & development , Animals , Breast/embryology , Breast/physiology , Female , Gene Expression Regulation, Developmental , Humans , In Vitro Techniques , Insulin-Like Growth Factor Binding Proteins/deficiency , Insulin-Like Growth Factor Binding Proteins/genetics , Mammary Glands, Animal/embryology , Mammary Glands, Animal/physiology , Mice , Mice, Knockout , Mice, Transgenic , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Somatomedins/physiology , Stromal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL