Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Pediatr Nephrol ; 34(3): 379-388, 2019 03.
Article in English | MEDLINE | ID: mdl-29214442

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) is the offending agent in post-diarrhea-associated hemolytic uremic syndrome (HUS), a disorder characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute kidney failure, with thrombi occluding the renal microvasculature. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Glomerular endothelial cells are susceptible to the toxic effects of Stxs that, via nuclear factor kappa B (NF-κB) activation, induce the expression of genes encoding for adhesion molecules and chemokines, culminating in leukocyte adhesion and platelet thrombus formation on the activated endothelium. Complement activation via the alternative pathway has been seen in patients during the acute phase of STEC-associated HUS. Experimental evidence has highlighted the role of complement proteins in driving glomerular endothelium toward a thrombogenic phenotype. At the glomerular level, podocytes are also an important target of Stx-induced complement activation. Glomerular injury as a consequence of podocyte dysfunction and loss is thus a mechanism that might affect long-term renal outcomes in the disease. New approaches to targeting the complement system may be useful therapeutic options for patients with STEC-HUS.


Subject(s)
Endothelial Cells/pathology , Hemolytic-Uremic Syndrome/immunology , Podocytes/pathology , Shiga Toxin/toxicity , Shiga-Toxigenic Escherichia coli/pathogenicity , Animals , Colon/microbiology , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology , Diarrhea/complications , Diarrhea/microbiology , Disease Models, Animal , Endothelial Cells/immunology , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Intestinal Mucosa/microbiology , Microvessels/cytology , Microvessels/immunology , Microvessels/pathology , Podocytes/immunology , Shiga Toxin/immunology , Shiga Toxin/metabolism , Shiga-Toxigenic Escherichia coli/immunology , Shiga-Toxigenic Escherichia coli/metabolism
2.
J Am Soc Nephrol ; 29(7): 1799-1809, 2018 07.
Article in English | MEDLINE | ID: mdl-29712732

ABSTRACT

Sirtuins belong to an evolutionarily conserved family of NAD+-dependent deacetylases that share multiple cellular functions related to proliferation, DNA repair, mitochondrial energy homeostasis, and antioxidant activity. Mammalians express seven sirtuins (SIRT1-7) that are localized in different subcellular compartments. Changes in sirtuin expression are critical in several diseases, including metabolic syndrome, diabetes, cancer, and aging. In the kidney, the most widely studied sirtuin is SIRT1, which exerts cytoprotective effects by inhibiting cell apoptosis, inflammation, and fibrosis together with SIRT3, a crucial metabolic sensor that regulates ATP generation and mitochondrial adaptive response to stress. Here, we provide an overview of the biologic effects of sirtuins and the molecular targets thereof regulating renal physiology. This review also details progress made in understanding the effect of sirtuins in the pathophysiology of chronic and acute kidney diseases, highlighting the key role of SIRT1, SIRT3, and now SIRT6 as potential therapeutic targets. In this context, the current pharmacologic approaches to enhancing the activity of SIRT1 and SIRT3 will be discussed.


Subject(s)
Acute Kidney Injury/metabolism , Aging/physiology , Kidney/metabolism , Kidney/pathology , Renal Insufficiency, Chronic/metabolism , Sirtuins/metabolism , Acute Kidney Injury/physiopathology , Animals , Diabetic Nephropathies/metabolism , Fibrosis , Humans , Kidney/physiology , Renal Insufficiency, Chronic/physiopathology , Renin-Angiotensin System , Sirtuin 1/metabolism
3.
Int J Mol Sci ; 20(11)2019 Jun 07.
Article in English | MEDLINE | ID: mdl-31181604

ABSTRACT

The prevalence of renal diseases is emerging as a public health problem. Despite major progress in supportive therapy, mortality rates among patients remain high. In an attempt to find innovative treatments to stimulate kidney regeneration, stem cell-based technology has been proposed as a potentially promising strategy. Here, we summarise the renoprotective potential of pluripotent and adult stem cell therapy in experimental models of acute and chronic kidney injury and we explore the different mechanisms at the basis of stem cell-induced kidney regeneration. Specifically, cell engraftment, incorporation into renal structures, or paracrine activities of embryonic or induced pluripotent stem cells as well as mesenchymal stem cells and renal precursors are analysed. We also discuss the relevance of stem cell secretome-derived bioproducts, including soluble factors and extracellular vesicles, and the option of using them as cell-free therapy to induce reparative processes. The translation of the experimental results into clinical trials is also addressed, highlighting the safety and feasibility of stem cell treatments in patients with kidney injury.


Subject(s)
Kidney Diseases/therapy , Stem Cell Transplantation/methods , Animals , Clinical Trials as Topic , Humans , Stem Cell Transplantation/adverse effects
4.
J Am Soc Nephrol ; 27(5): 1400-11, 2016 05.
Article in English | MEDLINE | ID: mdl-26516208

ABSTRACT

Generating kidney organoids using human stem cells could offer promising prospects for research and therapeutic purposes. However, no cell-based strategy has generated nephrons displaying an intact three-dimensional epithelial filtering barrier. Here, we generated organoids using murine embryonic kidney cells, and documented that these tissues recapitulated the complex three-dimensional filtering structure of glomerular slits in vivo and accomplished selective glomerular filtration and tubular reabsorption. Exploiting this technology, we mixed human amniotic fluid stem cells with mouse embryonic kidney cells to establish three-dimensional chimeric organoids that engrafted in vivo and grew to form vascularized glomeruli and tubular structures. Human cells contributed to the formation of glomerular structures, differentiated into podocytes with slit diaphragms, and internalized exogenously infused BSA, thus attaining in vivo degrees of specialization and function unprecedented for donor stem cells. In conclusion, human amniotic fluid stem cell chimeric organoids may offer new paths for studying renal development and human podocyte disease, and for facilitating drug discovery and translational research.


Subject(s)
Amniotic Fluid/cytology , Organoids/cytology , Podocytes , Stem Cells , Animals , Cells, Cultured , Humans , Kidney/cytology , Mice
5.
J Am Soc Nephrol ; 25(8): 1786-98, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24578132

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli is the offending agent of postdiarrhea-associated hemolytic uremic syndrome (HUS), a disorder of glomerular ischemic damage and widespread microvascular thrombosis. We previously documented that Stx induces glomerular complement activation, generating C3a responsible for microvascular thrombosis in experimental HUS. Here, we show that the presence of C3 deposits on podocytes is associated with podocyte damage and loss in HUS mice generated by the coinjection of Stx2 and LPS. Because podocyte adhesion to the glomerular basement membrane is mediated by integrins, the relevance of integrin-linked kinase (ILK) signals in podocyte dysfunction was evaluated. Podocyte expression of ILK increased after the injection of Stx2/LPS and preceded the upregulation of Snail and downregulation of nephrin and α-actinin-4. Factor B deficiency or pretreatment with an inhibitory antibody to factor B protected mice against Stx2/LPS-induced podocyte dysregulation. Similarly, pretreatment with a C3a receptor antagonist limited podocyte loss and changes in ILK, Snail, and α-actinin-4 expression. In cultured podocytes, treatment with C3a reduced α-actinin-4 expression and promoted ILK-dependent nuclear expression of Snail and cell motility. These results suggest that Stx-induced activation of the alternative pathway of complement and generation of C3a promotes ILK signaling, leading to podocyte dysfunction and loss in Stx-HUS.


Subject(s)
Complement C3a/metabolism , Complement Pathway, Alternative/drug effects , Hemolytic-Uremic Syndrome/pathology , Kidney Glomerulus/drug effects , Podocytes/drug effects , Shiga Toxin 2/pharmacology , Animals , Cell Culture Techniques , Disease Models, Animal , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/metabolism , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Podocytes/metabolism , Podocytes/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
6.
J Am Soc Nephrol ; 25(3): 523-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371298

ABSTRACT

Activation of endothelin-A receptor (ET(A)R) by endothelin-1 (ET-1) drives epithelial-to-mesenchymal transition in ovarian tumor cells through ß-arrestin signaling. Here, we investigated whether this pathogenetic pathway could affect podocyte phenotype in proliferative glomerular disorders. In cultured mouse podocytes, ET-1 caused loss of the podocyte differentiation marker synaptopodin and acquisition of the mesenchymal marker α-smooth muscle actin. ET-1 promoted podocyte migration via ET(A)R activation and increased ß-arrestin-1 expression. Activated ET(A)R recruited ß-arrestin-1 to form a trimeric complex with Src leading to epithelial growth factor receptor (EGFR) transactivation and ß-catenin phosphorylation, which promoted gene transcription of Snail. Increased Snail expression fostered ET-1-induced migration as confirmed by Snail knockdown experiments. Silencing of ß-arrestin-1 prevented podocyte phenotypic changes and motility and inhibited ET(A)R-driven signaling. In vitro findings were confirmed in doxorubicin (Adriamycin)-induced nephropathy. Mice receiving Adriamycin developed renal injury with loss of podocytes and hyperplastic lesion formation; ß-arrestin-1 expression increased in visceral podocytes and in podocytes entrapped in pseudo-crescents. Administration of the selective ET(A)R antagonist sitaxsentan prevented podocyte loss, formation of the hyperplastic lesions, and normalized expression of glomerular ß-arrestin-1 and Snail. Increased ß-arrestin-1 levels in podocytes retrieved from crescents of patients with proliferative glomerulopathies confirmed the translational relevance of these findings and suggest the therapeutic potential of ET(A)R antagonism for a group of diseases still needing a specific treatment.


Subject(s)
Arrestins/physiology , Endothelin-1/metabolism , Glomerulonephritis/chemically induced , Podocytes/physiology , Receptor, Endothelin A/metabolism , Animals , Cell Movement , Disease Models, Animal , Doxorubicin , ErbB Receptors/metabolism , Female , Glomerulonephritis/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Snail Family Transcription Factors , Transcription Factors/metabolism , Transcriptional Activation , beta Catenin/metabolism , beta-Arrestin 1 , beta-Arrestins , src-Family Kinases/metabolism
7.
N Engl J Med ; 365(4): 295-306, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21756023

ABSTRACT

BACKGROUND: Focal segmental glomerulosclerosis is a kidney disease that is manifested as the nephrotic syndrome. It is often resistant to glucocorticoid therapy and progresses to end-stage renal disease in 50 to 70% of patients. Genetic studies have shown that familial focal segmental glomerulosclerosis is a disease of the podocytes, which are major components of the glomerular filtration barrier. However, the molecular cause in over half the cases of primary focal segmental glomerulosclerosis is unknown, and effective treatments have been elusive. METHODS: We performed whole-genome linkage analysis followed by high-throughput sequencing of the positive-linkage area in a family with autosomal recessive focal segmental glomerulosclerosis (index family) and sequenced a newly discovered gene in 52 unrelated patients with focal segmental glomerulosclerosis. Immunohistochemical studies were performed on human kidney-biopsy specimens and cultured podocytes. Expression studies in vitro were performed to characterize the functional consequences of the mutations identified. RESULTS: We identified two mutations (A159P and Y695X) in MYO1E, which encodes a nonmuscle class I myosin, myosin 1E (Myo1E). The mutations in MYO1E segregated with focal segmental glomerulosclerosis in two independent pedigrees (the index family and Family 2). Patients were homozygous for the mutations and did not have a response to glucocorticoid therapy. Electron microscopy showed thickening and disorganization of the glomerular basement membrane. Normal expression of Myo1E was documented in control human kidney-biopsy specimens in vivo and in glomerular podocytes in vitro. Transfection studies revealed abnormal subcellular localization and function of the A159P-Myo1E mutant. The Y695X mutation causes loss of calmodulin binding and of the tail domains of Myo1E. CONCLUSIONS: MYO1E mutations are associated with childhood-onset, glucocorticoid-resistant focal segmental glomerulosclerosis. Our data provide evidence of a role of Myo1E in podocyte function and the consequent integrity of the glomerular filtration barrier.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Mutation , Myosin Type I/genetics , Age of Onset , Animals , Child , Child, Preschool , Drug Resistance , Female , Genes, Recessive , Genetic Linkage , Genome-Wide Association Study , Glomerulosclerosis, Focal Segmental/pathology , Glucocorticoids/therapeutic use , Humans , Infant , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Male , Mice , Microscopy, Fluorescence , Mutation, Missense , Myosin Type I/chemistry , Myosin Type I/metabolism , Pedigree , Podocytes/metabolism , Podocytes/ultrastructure , Sequence Alignment
8.
Am J Pathol ; 183(1): 119-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23707238

ABSTRACT

In nondiabetic rat models of renal disease, angiotensin II (Ang II) perpetuates podocyte injury and promotes progression to end-stage kidney disease. Herein, we wanted to explore the role of Ang II in diabetic nephropathy by a translational approach spanning from in vitro to in vivo rat and human studies, and to dissect the intracellular pathways involved. In isolated perfused rat kidneys and in cultured human podocytes, Ang II down-regulated nephrin expression via Notch1 activation and nuclear translocation of Snail. Hairy enhancer of split-1 was a Notch1-downstream gene effector that activated Snail in cultured podocytes. In vitro changes of the Snail/nephrin axis were similar to those in renal biopsy specimens of Zucker diabetic fatty rats and patients with advanced diabetic nephropathy, and were normalized by pharmacological inhibition of the renin-angiotensin system. Collectively, the present studies provide evidence that Ang II plays a relevant role in perpetuating glomerular injury in experimental and human diabetic nephropathy via persistent activation of Notch1 and Snail signaling in podocytes, eventually resulting in down-regulation of nephrin expression, the integrity of which is crucial for the glomerular filtration barrier.


Subject(s)
Angiotensin II/metabolism , Diabetic Nephropathies/metabolism , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Transcription Factors/metabolism , Aged , Animals , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Down-Regulation , Female , Humans , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Linear Models , Male , Microscopy, Electron, Transmission , Middle Aged , Rats , Rats, Sprague-Dawley , Rats, Zucker , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors
9.
Nephron Exp Nephrol ; 126(2): 59, 2014.
Article in English | MEDLINE | ID: mdl-24854642

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is emerging as a public health problem in developing and developed countries. It affects up to 7% of hospitalized patients, with a higher prevalence in critical care units. Despite major advances in preventive strategies and support measures, the mortality rate among patients remains higher than 50%. Several pharmacological approaches to improve renal function and survival after an AKI episode have been largely unsuccessful in clinical practice. SUMMARY: Stem cell-based therapy has provided new hopes of innovative interventions to enhance the limited capability of kidney regeneration in AKI. An important target for cell therapy is represented by tubular epithelial cells which after acute ischemic or toxic insults undergo dysfunction and detachment. Among adult stem cells, mesenchymal stromal/stem cells (MSC) are an attractive therapeutic tool by virtue of their unique biological properties, tropism for damaged tissues, and proregenerative capacity. In the present review, we discuss the mechanisms underlying the renoprotective effects of therapies with stem cells of different origins in preclinical models of AKI by evaluating new modalities by which MSC interact with damaged cells via the release of soluble factors and exosomes/microvesicles. Several biological effects, including antiapoptotic, promitogenic, immunomodulatory, and anti-inflammatory activities, have been analyzed in renal tissue of AKI animals receiving stem cell treatments. The mechanisms of stem cell homing and engraftment to sites of tissue damage have also been discussed. KEY MESSAGES: The translation of preclinical data on stem cells into effective and safe new modalities of care is still limited, and further studies are needed before their application in patients with AKI.


Subject(s)
Acute Kidney Injury/therapy , Amniotic Fluid/cytology , Mesenchymal Stem Cell Transplantation , Stem Cell Transplantation , Animals , Humans
10.
Nephron ; 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38452744

ABSTRACT

The complement system, a cornerstone of the innate immune defense, typically confers protection against pathogens. However, in various clinical scenarios the complement's defensive actions can harm host cells, exacerbating immune and inflammatory responses. The central components C3 and C5 undergo proteolytic cleavage during complement activation, yielding small active fragments C3a and C5a anaphylatoxins. Traditionally these fragments were associated with inflammation via the specific receptors C3a receptor (R), C5aR1 and C5aR2. Recent insights, however, spotlight the excessive C3a/C3aR and C5a/C5aR1 signaling as culprits in diverse disorders of inflammatory and autoimmune etiology. This is particularly true for several kidney diseases, where the potential involvement of anaphylatoxins in renal damage is supported by the enhanced renal expression of their receptors and the high levels of C3a and C5a in both plasma and urine. Furthermore, the production of complement proteins in the kidney, with different renal cells synthesizing C3 and C5, significantly contributes to local tissue injury. In the present review, we discuss the different aspects of C3a/C3aR and C5a/C5aR signaling in acute and chronic kidney diseases, and explore the therapeutic potential of emerging targeted drugs for future clinical applications.

11.
Nephrol Dial Transplant ; 28(4): 788-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23258756

ABSTRACT

Acute renal failure (ARF; acute kidney injury-according to the more recent classification) is emerging as a public health problem. Despite major advances in supportive therapy, the mortality and morbidity among patients remain dismally high. In the attempt to yield innovative interventions fostering the limited capability of regeneration of the kidney, several studies have tested stem cell-based technology mainly employing mesenchymal stem cells (MSC) of different origins. The results of this approach provide the exciting prospect of a powerful treatment to repair acutely damaged organs by virtue of the unique MSC tropism for the damaged tissue, as well as their paracrine action. In the present review, we discuss the mechanisms underlying the regenerative processes triggered by MSC therapy in preclinical models of ARF by analysing modalities of cell-to-cell communication through the release of soluble factors and microvesicles/exosomes by MSC into the damaged renal tissue. Key receptors involved in MSC homing, engraftment and survival at the sites of injury are also elucidated. A translation of basic discoveries of MSC biology into effective care is still limited to the preliminary data of a phase I clinical trial, and further studies are needed to definitively assess the efficacy of MSC-based therapy in humans.


Subject(s)
Acute Kidney Injury/prevention & control , Mesenchymal Stem Cells , Animals , Humans
12.
J Immunol ; 187(1): 172-80, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21642543

ABSTRACT

Shiga toxin (Stx)-producing E.coli O157:H7 has become a global threat to public health; it is a primary cause of diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure with thrombi occluding renal microcirculation. In this study, we explored whether Stx triggers complement-dependent microvascular thrombosis in in vitro and in vivo experimental settings of HUS. Stx induced on human microvascular endothelial cell surface the expression of P-selectin, which bound and activated C3 via the alternative pathway, leading to thrombus formation under flow. In the search for mechanisms linking complement activation and thrombosis, we found that exuberant complement activation in response to Stx generated an increased amount of C3a that caused further endothelial P-selectin expression, thrombomodulin (TM) loss, and thrombus formation. In a murine model of HUS obtained by coinjection of Stx2 and LPS and characterized by thrombocytopenia and renal dysfunction, upregulation of glomerular endothelial P-selectin was associated with C3 and fibrin(ogen) deposits, platelet clumps, and reduced TM expression. Treatment with anti-P-selectin Ab limited glomerular C3 accumulation. Factor B-deficient mice after Stx2/LPS exhibited less thrombocytopenia and were protected against glomerular abnormalities and renal function impairment, indicating the involvement of complement activation via the alternative pathway in the glomerular thrombotic process in HUS mice. The functional role of C3a was documented by data showing that glomerular fibrin(ogen), platelet clumps, and TM loss were markedly decreased in HUS mice receiving C3aR antagonist. These results identify Stx-induced complement activation, via P-selectin, as a key mechanism of C3a-dependent microvascular thrombosis in diarrhea-associated HUS.


Subject(s)
Complement C3a/toxicity , Complement Pathway, Alternative/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Animals , Cell Line , Complement C3a/biosynthesis , Complement C3a/metabolism , Complement Factor B/deficiency , Complement Factor B/genetics , Disease Models, Animal , Endothelium, Vascular/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Hemolytic-Uremic Syndrome/metabolism , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/immunology , P-Selectin/physiology , Protein Binding/immunology
13.
J Immunol ; 186(7): 4164-74, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21389254

ABSTRACT

Tolerance induction toward allogeneic organ grafts represents one of the major aims of transplantation medicine. Stem cells are promising candidates for promoting donor-specific tolerance. In this study, we investigated the immunomodulatory properties of murine embryonic stem cells (ESCs), obtained either by in vitro fertilization (IVF-ESCs) or by nuclear transfer (NT-ESCs), in heart transplant mouse models. IVF-ESCs did not prolong the survival of fully allogeneic cardiac transplants but significantly prolonged the survival of semiallogeneic hearts from the same ESC donor strain for >100 d in 44% of the animals. However, 28% of transplanted animals infused with IVF-ESCs experienced development of a teratoma. NT-ESCs similarly prolonged semiallogeneic heart graft survival (>100 d in 40% of the animals) but were less teratogenic. By in vitro studies, IVF-ESC and NT-ESC immunoregulation was mediated both by cell contact-dependent mechanisms and by the release of soluble factors. By adding specific inhibitors, we identified PGE(2) as a soluble mediator of ESC immunoregulation. Expansion of regulatory T cells was found in lymphoid organs and in the grafts of IVF-ESC- and NT-ESC-tolerized mice. Our study demonstrates that both IVF-ESCs and NT-ESCs modulate recipient immune response toward tolerance to solid organ transplantation, and that NT-ESCs exhibit a lower tendency for teratoma formation. Because NT-ESCs are obtained by NT of a somatic cell from living individuals into an enucleated oocyte, they could represent a source of donor-derived stem cells to induce tolerance to solid organ allograft.


Subject(s)
Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Fertilization in Vitro , Graft Survival/immunology , Heart Transplantation/immunology , Nuclear Proteins/administration & dosage , Adoptive Transfer , Animals , Cell Line , Female , Graft Rejection/prevention & control , Heart Transplantation/methods , Heart Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Random Allocation , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
14.
J Am Soc Nephrol ; 23(11): 1857-68, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23085631

ABSTRACT

The shortage of transplantable organs provides an impetus to develop tissue-engineered alternatives. Producing tissues similar to immature kidneys from simple suspensions of fully dissociated embryonic renal cells is possible in vitro, but glomeruli do not form in the avascular environment. Here, we constructed renal organoids from single-cell suspensions derived from E11.5 kidneys and then implanted these organoids below the kidney capsule of a living rat host. This implantation resulted in further maturation of kidney tissue, formation of vascularized glomeruli with fully differentiated capillary walls, including the slit diaphragm, and appearance of erythropoietin-producing cells. The implanted tissue exhibited physiologic functions, including tubular reabsorption of macromolecules, that gained access to the tubular lumen on glomerular filtration. The ability to generate vascularized nephrons from single-cell suspensions marks a significant step to the long-term goal of replacing renal function by a tissue-engineered kidney.


Subject(s)
Kidney/embryology , Organoids/embryology , Tissue Engineering/methods , Animals , Bioartificial Organs , Kidney/blood supply , Kidney/cytology , Kidney/physiology , Kidney Transplantation/methods , Male , Mice , Organoids/cytology , Organoids/transplantation , Rats , Rats, Nude , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/administration & dosage
15.
PLoS One ; 18(10): e0291909, 2023.
Article in English | MEDLINE | ID: mdl-37816025

ABSTRACT

Sirtuin 3 (SIRT3), the main deacetylase of mitochondria, modulates the acetylation levels of substrates governing metabolism and oxidative stress. In the kidney, we showed that SIRT3 affects the proper functioning of high energy-demanding cells, such as tubular cells and podocytes. Less is known about the role of SIRT3 in regulating endothelial cell function and its impact on the progression of kidney disease. Here, we found that whole body Sirt3-deficient mice exhibited reduced renal capillary density, reflecting endothelial dysfunction, and VEGFA expression compared to wild-type mice. This was paralleled by activation of hypoxia signaling, upregulation of HIF-1α and Angiopietin-2, and oxidative stress increase. These alterations did not result in kidney disease. However, when Sirt3-deficient mice were exposed to the nephrotoxic stimulus Adriamycin (ADR) they developed aggravated endothelial rarefaction, altered VEGFA signaling, and higher oxidative stress compared to wild-type mice receiving ADR. As a result, ADR-treated Sirt3-deficient mice experienced a more severe injury with exacerbated albuminuria, podocyte loss and fibrotic lesions. These data suggest that SIRT3 is a crucial regulator of renal vascular homeostasis and its dysregulation is a predisposing factor for kidney disease. By extension, our findings indicate SIRT3 as a pharmacologic target in progressive renal disease whose treatments are still imperfect.


Subject(s)
Kidney Diseases , Sirtuin 3 , Vascular Diseases , Mice , Animals , Sirtuin 3/metabolism , Kidney/metabolism , Oxidative Stress , Kidney Diseases/genetics , Kidney Diseases/metabolism , Mitochondria/metabolism , Vascular Diseases/metabolism
16.
Sci Rep ; 13(1): 11392, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37452090

ABSTRACT

The spike protein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) can interact with endothelial cells. However, no studies demonstrated the direct effect of the spike protein subunit 1 (S1) in inducing lung vascular damage and the potential mechanisms contributing to lung injury. Here, we found that S1 injection in mice transgenic for human angiotensin converting enzyme 2 (ACE2) induced early loss of lung endothelial thromboresistance at 3 days, as revealed by thrombomodulin loss and von Willebrand factor (vWF) increase. In parallel, vascular and epithelial C3 deposits and enhanced C3a receptor (C3aR) expression were observed. These changes preceded diffuse alveolar damage and lung vascular fibrin(ogen)/platelets aggregates at 7 days, as well as inflammatory cell recruitment and fibrosis. Treatment with C3aR antagonist (C3aRa) inhibited lung C3 accumulation and C3a/C3aR activation, limiting vascular thrombo-inflammation and fibrosis. Our study demonstrates that S1 triggers vascular dysfunction and activates complement system, instrumental to lung thrombo-inflammatory injury. By extension, our data indicate C3aRa as a valuable therapeutic strategy to limit S1-dependent lung pathology.


Subject(s)
Complement C3a , Endothelial Cells , Receptors, Complement , Spike Glycoprotein, Coronavirus , SARS-CoV-2 , Endothelial Cells/cytology , Endothelial Cells/virology , Lung/pathology , Lung/virology , Complement C3a/metabolism , Receptors, Complement/metabolism , Fibrosis , Mice, Transgenic , Humans , Animals , Mice , COVID-19 , Inflammation
17.
Am J Physiol Renal Physiol ; 303(9): F1370-81, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22952284

ABSTRACT

We previously reported that in a model of spontaneously progressive glomerular injury with early podocyte loss, abnormal migration, and proliferation of glomerular parietal epithelial progenitor cells contributed to the formation of synechiae and crescentic lesions. Here we first investigated whether a similar sequence of events could be extended to rats with adriamycin (ADR)-induced nephropathy. As a second aim, the regenerative potential of therapy with bone marrow-derived mesenchymal stem cells (MSCs) on glomerular resident cells was evaluated. In ADR-treated rats, decrease of WT1(+) podocyte number due to apoptosis was associated with reduced glomerular expression of nephrin and CD2AP. As a consequence of podocyte injury, glomerular adhesions of the capillary tuft to the Bowman's capsule were observed, followed by crescent-like lesions and glomerulosclerosis. Cellular components of synechiae were either NCAM(+) parietal progenitor cells or nestin(+) podocytes. In ADR rats, repeated injections of MSCs limited podocyte loss and apoptosis and partially preserved nephrin and CD2AP. MSCs attenuated the formation of glomerular podocyte-parietal epithelial cell bridges and normalized the distribution of NCAM(+) progenitor cells along the Bowman's capsule, thereby reducing glomerulosclerosis. Finding that MSCs increased glomerular VEGF expression and limited microvascular rarefaction may explain the prosurvival effect by stem cell therapy. MSCs also displayed anti-inflammatory activity. Coculture of MSCs with ADR-damaged podocytes showed a functional role of stem cell-derived VEGF on prosurvival pathways. These data suggest that MSCs by virtue of their tropism for damaged kidney and ability to provide a local prosurvival environment may represent a useful strategy to preserve podocyte viability and reduce glomerular inflammation and sclerosis.


Subject(s)
Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney/physiology , Mesenchymal Stem Cell Transplantation , Podocytes/physiology , Regeneration/physiology , Stem Cells/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/physiology , Cell Count , Cell Movement/physiology , Coculture Techniques , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Doxorubicin/adverse effects , Kidney/pathology , Kidney Diseases/chemically induced , Male , Membrane Proteins/metabolism , Podocytes/pathology , Rats , Rats, Inbred Lew , Stem Cells/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
18.
Am J Pathol ; 179(2): 628-38, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21718676

ABSTRACT

We previously reported that angiotensin-converting enzyme inhibitor (ACEi) renoprotection in Munich Wistar Frömter (MWF) rats, which develop progressive glomerular injury, was associated with podocyte repopulation and preservation of glomerular architecture. Here, we studied the time course of the lesions, their cellular components, and the effect of ACEi. Early glomerular lesions were synechiae, followed by extracapillary crescents and glomerulosclerosis. The majority of cells forming crescents were claudin1(+) parietal epithelial cells and, to a lesser extent, WT1(+) podocytes, both in active proliferation. In crescents, cells expressing the metanephric mesenchyme marker NCAM were also found. Three distinct populations of parietal epithelial cells were identified in the rat Bowman's capsule: NCAM(+)WT1(-) cells, also expressing progenitor cell marker CD24, and NCAM(+)WT1(+) and NCAM(-)WT1(+) cells, the latter population representing parietal podocytes. After exposure to inductive medium, cultured parietal epithelial cells that were obtained by capsulated glomeruli generated podocytes, documenting their progenitor nature. Mitotic activity of cultured renal progenitors was induced by angiotensin II through the down-regulation of cell cycle inhibitor C/EBPδ expression. Treatment with ACEi reduced number and extension of crescents and glomerulosclerosis in MWF rats. Renoprotection was accomplished through the limitation of NCAM(+) progenitor proliferation via the modulation of C/EBPδ. Thus, chaotic migration and proliferation of the Bowman's capsule progenitor cells pave the way to crescent formation and subsequent sclerosis. ACEi, by moderating progenitor cell activation, restores glomerular architecture and prevents renal disease progression.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney Glomerulus/enzymology , Kidney/enzymology , Animals , Cell Proliferation , Creatinine/blood , Humans , Kidney/pathology , Kidney Failure, Chronic/metabolism , Kidney Glomerulus/pathology , Male , Microscopy, Immunoelectron/methods , Mitosis , Podocytes/cytology , Rats , Rats, Wistar , Stem Cells/cytology , Time Factors
19.
Front Immunol ; 13: 827146, 2022.
Article in English | MEDLINE | ID: mdl-35320941

ABSTRACT

Microvascular thrombosis is associated with multiorgan failure and mortality in coronavirus disease 2019 (COVID-19). Although thrombotic complications may be ascribed to the ability of SARS-CoV-2 to infect and replicate in endothelial cells, it has been poorly investigated whether, in the complexity of viral infection in the human host, specific viral elements alone can induce endothelial damage. Detection of circulating spike protein in the sera of severe COVID-19 patients was evaluated by ELISA. In vitro experiments were performed on human microvascular endothelial cells from the derma and lung exposed to SARS-CoV-2-derived spike protein 1 (S1). The expression of adhesive molecules was studied by immunofluorescence and leukocyte adhesion and platelet aggregation were assessed under flow conditions. Angiotensin converting enzyme 2 (ACE2) and AMPK expression were investigated by Western Blot analysis. In addition, S1-treated endothelial cells were incubated with anti-ACE2 blocking antibody, AMPK agonist, or complement inhibitors. Our results show that significant levels of spike protein were found in the 30.4% of severe COVID-19 patients. In vitro, the activation of endothelial cells with S1 protein, via ACE2, impaired AMPK signalling, leading to robust leukocyte recruitment due to increased adhesive molecule expression and thrombomodulin loss. This S1-induced pro-inflammatory phenotype led to exuberant C3 and C5b-9 deposition on endothelial cells, along with C3a and C5a generation that further amplified S1-induced complement activation. Functional blockade of ACE2 or complement inhibition halted S1-induced platelet aggregates by limiting von Willebrand factor and P-selectin exocytosis and expression on endothelial cells. Overall, we demonstrate that SARS-CoV-2-derived S1 is sufficient in itself to propagate inflammatory and thrombogenic processes in the microvasculature, amplified by the complement system, recapitulating the thromboembolic complications of COVID-19.


Subject(s)
COVID-19 , Spike Glycoprotein, Coronavirus , AMP-Activated Protein Kinases/metabolism , Angiotensin-Converting Enzyme 2 , Complement System Proteins/metabolism , Endothelial Cells/metabolism , Humans , Platelet Aggregation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism
20.
Cells ; 11(20)2022 10 21.
Article in English | MEDLINE | ID: mdl-36291179

ABSTRACT

A reduced nephron number at birth, due to critical gestational conditions, including maternal malnutrition, is associated with the risk of developing hypertension and chronic kidney disease in adulthood. No interventions are currently available to augment nephron number. We have recently shown that sirtuin 3 (SIRT3) has an important role in dictating proper nephron endowment. The present study explored whether SIRT3 stimulation, by means of supplementation with nicotinamide riboside (NR), a precursor of the SIRT3 co-substrate nicotinamide adenine dinucleotide (NAD+), was able to improve nephron number in a murine model of a low protein (LP) diet. Our findings show that reduced nephron number in newborn mice (day 1) born to mothers fed a LP diet was associated with impaired renal SIRT3 expression, which was restored through supplementation with NR. Glomerular podocyte density, as well as the rarefaction of renal capillaries, also improved through NR administration. In mechanistic terms, the restoration of SIRT3 expression through NR was mediated by the induction of proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α). Moreover, NR restored SIRT3 activity, as shown by the reduction of the acetylation of optic atrophy 1 (OPA1) and superoxide dismutase 2 (SOD2), which resulted in improved mitochondrial morphology and protection against oxidative damage in mice born to mothers fed the LP diet. Our results provide evidence that it is feasible to prevent nephron mass shortage at birth through SIRT3 boosting during nephrogenesis, thus providing a therapeutic option to possibly limit the long-term sequelae of reduced nephron number in adulthood.


Subject(s)
Sirtuin 3 , Mice , Animals , Sirtuin 3/metabolism , NAD , Diet, Protein-Restricted , PPAR gamma , Nephrons/metabolism , Dietary Supplements
SELECTION OF CITATIONS
SEARCH DETAIL