Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 20(11)2019 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-31181604

RESUMO

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.


Assuntos
Nefropatias/terapia , Transplante de Células-Tronco/métodos , Animais , Ensaios Clínicos como Assunto , Humanos , Transplante de Células-Tronco/efeitos adversos
2.
Curr Opin Organ Transplant ; 20(1): 86-93, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25522138

RESUMO

PURPOSE OF REVIEW: Ongoing research is constantly looking for means to modulate the immune system for long-lasting engraftment of pluripotent stem cells (PSC) during stem cell-based therapies. This study reviews data on in-vitro and in-vivo immunogenicity of embryonic and induced-PSC and describes how their immunological properties can be harnessed for tolerance induction in organ transplantation. RECENT FINDINGS: Although PSC display immunomodulatory properties in vitro, they are capable of eliciting an immune response that leads to cell rejection when transplanted into immune-competent recipients. Nevertheless, long-term acceptance of PSC-derived cells/tissues in an allogeneic environment can be achieved using minimal host conditioning. Protocols for differentiating PSC towards haematopoietic stem cells, thymic epithelial precursors, dendritic cells, regulatory T cells and myeloid-derived suppressor cells are being developed, suggesting the possibility to use PSC-derived immunomodulatory cells to induce tolerance to a solid organ transplant. SUMMARY: PSC and/or their derivatives possess unique immunological properties that allow for acceptance of PSC-derived tissue with minimal host conditioning. Investigators involved either in regenerative or in transplant medicine must join their efforts with the ultimate aim of using PSC as a source of donor-specific cells that would create a protolerogenic environment to achieve tolerance in solid organ transplantation.


Assuntos
Imunidade Adaptativa/imunologia , Tolerância Imunológica/imunologia , Transplante de Órgãos , Células-Tronco Pluripotentes/imunologia , Transplante de Células-Tronco , Animais , Humanos
3.
Nephron ; 148(10): 712-723, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38452744

RESUMO

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.


Assuntos
Complemento C3a , Complemento C5a , Inflamação , Transdução de Sinais , Humanos , Complemento C5a/metabolismo , Complemento C5a/imunologia , Complemento C3a/metabolismo , Inflamação/metabolismo , Animais , Nefropatias/metabolismo , Nefropatias/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Receptores de Complemento/metabolismo
4.
J Immunol ; 186(7): 4164-74, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21389254

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/transplante , Fertilização in vitro , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Proteínas Nucleares/administração & dosagem , Transferência Adotiva , Animais , Linhagem Celular , Feminino , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/métodos , Transplante de Coração/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Distribuição Aleatória , Transplante Homólogo/imunologia , Transplante Homólogo/patologia
5.
PLoS One ; 18(10): e0291909, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37816025

RESUMO

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.


Assuntos
Nefropatias , Sirtuína 3 , Doenças Vasculares , Camundongos , Animais , Sirtuína 3/metabolismo , Rim/metabolismo , Estresse Oxidativo , Nefropatias/genética , Nefropatias/metabolismo , Mitocôndrias/metabolismo , Doenças Vasculares/metabolismo
6.
Sci Rep ; 13(1): 11392, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37452090

RESUMO

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.


Assuntos
Complemento C3a , Células Endoteliais , Receptores de Complemento , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2 , Células Endoteliais/citologia , Células Endoteliais/virologia , Pulmão/patologia , Pulmão/virologia , Complemento C3a/metabolismo , Receptores de Complemento/metabolismo , Fibrose , Camundongos Transgênicos , Humanos , Animais , Camundongos , COVID-19 , Inflamação
7.
Front Immunol ; 13: 827146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35320941

RESUMO

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.


Assuntos
COVID-19 , Glicoproteína da Espícula de Coronavírus , Proteínas Quinases Ativadas por AMP/metabolismo , Enzima de Conversão de Angiotensina 2 , Proteínas do Sistema Complemento/metabolismo , Células Endoteliais/metabolismo , Humanos , Agregação Plaquetária , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo
8.
Cells ; 11(20)2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36291179

RESUMO

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.


Assuntos
Sirtuína 3 , Camundongos , Animais , Sirtuína 3/metabolismo , NAD , Dieta com Restrição de Proteínas , PPAR gama , Néfrons/metabolismo , Suplementos Nutricionais
9.
Cells ; 11(11)2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35681450

RESUMO

Shiga toxin (Stx)-producing Escherichia coli is the predominant offending agent of post-diarrheal hemolytic uremic syndrome (HUS), a rare disorder of microvascular thrombosis and acute kidney injury possibly leading to long-term renal sequelae. We previously showed that C3a has a critical role in the development of glomerular damage in experimental HUS. Based on the evidence that activation of C3a/C3a receptor (C3aR) signaling induces mitochondrial dysregulation and cell injury, here we investigated whether C3a caused podocyte and tubular injury through induction of mitochondrial dysfunction in a mouse model of HUS. Mice coinjected with Stx2/LPS exhibited glomerular podocyte and tubular C3 deposits and C3aR overexpression associated with cell damage, which were limited by C3aR antagonist treatment. C3a promoted renal injury by affecting mitochondrial wellness as demonstrated by data showing that C3aR blockade reduced mitochondrial ultrastructural abnormalities and preserved mitochondrial mass and energy production. In cultured podocytes and tubular cells, C3a caused altered mitochondrial fragmentation and distribution, and reduced anti-oxidant SOD2 activity. Stx2 potentiated the responsiveness of renal cells to the detrimental effects of C3a through increased C3aR protein expression. These results indicate that C3aR may represent a novel target in Stx-associated HUS for the preservation of renal cell integrity through the maintenance of mitochondrial function.


Assuntos
Síndrome Hemolítico-Urêmica , Podócitos , Receptores de Complemento , Toxina Shiga II , Animais , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/metabolismo , Glomérulos Renais , Camundongos , Mitocôndrias/metabolismo , Podócitos/metabolismo , Receptores de Complemento/metabolismo , Toxina Shiga II/farmacologia
10.
Stem Cells ; 28(3): 513-22, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20049901

RESUMO

In search for new sources of mesenchymal stem cells (MSCs) for renal repair in acute kidney injury (AKI), we investigated the potential of human cord blood (CB)-MSCs to cure mice with AKI. Infusion of CB-MSCs in immunodeficient mice with cisplatin-induced AKI ameliorated both renal function and tubular cell injury, and prolonged survival. Transplanted CB-MSCs localized in peritubular areas, limited capillary alterations and neutrophil infiltration. Apoptosis reduced and tubular cell proliferation increased by virtue of stem cell capacity to produce growth factors. The reno-protective effect of CB-MSCs was further confirmed by their ability to inhibit oxidative damage and to induce the prosurvival factor Akt in tubular cells. The evidence that CB-MSCs in vitro increased the production of growth factors and inhibited IL-1 beta and TNFalpha synthesis when cocultured with damaged proximal tubular cells indicates a regenerative and anti-inflammatory action of stem cell treatment. Altogether these results highlight the potential of human CB-MSCs as future cell therapy for testing in human AKI.


Assuntos
Diferenciação Celular/fisiologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sobrevivência de Enxerto/fisiologia , Nefropatias/cirurgia , Rim/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Doença Aguda , Animais , Anti-Inflamatórios/metabolismo , Apoptose/fisiologia , Técnicas de Cultura de Células , Proliferação de Células , Técnicas de Cocultura , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/patologia , Rim/fisiopatologia , Nefropatias/fisiopatologia , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
Sci Rep ; 11(1): 23580, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34880332

RESUMO

Abnormal kidney development leads to lower nephron number, predisposing to renal diseases in adulthood. In embryonic kidneys, nephron endowment is dictated by the availability of nephron progenitors, whose self-renewal and differentiation require a relatively repressed chromatin state. More recently, NAD+-dependent deacetylase sirtuins (SIRTs) have emerged as possible regulators that link epigenetic processes to the metabolism. Here, we discovered a novel role for the NAD+-dependent deacylase SIRT3 in kidney development. In the embryonic kidney, SIRT3 was highly expressed only as a short isoform, with nuclear and extra-nuclear localisation. The nuclear SIRT3 did not act as deacetylase but exerted de-2-hydroxyisobutyrylase activity on lysine residues of histone proteins. Extra-nuclear SIRT3 regulated lysine 2-hydroxyisobutyrylation (Khib) levels of phosphofructokinase (PFK) and Sirt3 deficiency increased PFK Khib levels, inducing a glycolysis boost. This altered Khib landscape in Sirt3-/- metanephroi was associated with decreased nephron progenitors, impaired nephrogenesis and a reduced number of nephrons. These data describe an unprecedented role of SIRT3 in controlling early renal development through the regulation of epigenetics and metabolic processes.


Assuntos
Glicólise/genética , Nefropatias/genética , Organogênese/genética , Processamento de Proteína Pós-Traducional/genética , Sirtuína 3/genética , Animais , Diferenciação Celular/genética , Núcleo Celular/genética , Cromatina/genética , Epigênese Genética/genética , Rim/fisiologia , Lisina/genética , Camundongos , Camundongos Endogâmicos C57BL , NAD/genética , Néfrons/fisiologia , Fosfofrutoquinases/genética
12.
J Immunol ; 181(6): 3933-46, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768848

RESUMO

In this study, we investigated whether mesenchymal stem cells (MSC) had immunomodulatory properties in solid organ allotransplantation, using a semiallogeneic heart transplant mouse model, and studied the mechanism(s) underlying MSC tolerogenic effects. Either single (portal vein, day -7) or double (portal vein, day -7 and tail vein, day -1) pretransplant infusions of donor-derived B6C3 MSC in B6 recipients induced a profound T cell hyporesponsiveness and prolonged B6C3 cardiac allograft survival. The protolerogenic effect was abrogated when donor-derived MSC were injected together with B6C3 hematopoietic stem cells (HSC), suggesting that HSC negatively impact MSC immunomodulatory properties. Both the induction (pretransplant) and the maintenance phase (>100 days posttransplant) of donor-derived MSC-induced tolerance were associated with CD4(+)CD25(+)Foxp3(+) Treg expansion and impaired anti-donor Th1 activity. MSC-induced regulatory T cells (Treg) were donor-specific since adoptive transfer of splenocytes from tolerant mice prevented the rejection of fully MHC-mismatched donor-specific secondary allografts but not of third-party grafts. In addition, infusion of recipient-derived B6 MSC tolerized a semiallogeneic B6C3 cardiac allograft, but not a fully MHC-mismatched BALB/c graft, and expanded Treg. A double i.v. pretransplant infusion of recipient-derived MSC had the same tolerogenic effect as the combined intraportal/i.v. MSC infusions, which makes the tolerogenic protocol applicable in a clinical setting. In contrast, single MSC infusions given either peritransplant or 1 day after transplant were less effective. Altogether these findings indicate that MSC immunomodulatory properties require HSC removal, partial sharing of MHC Ags between the donor and the recipient and pretransplant infusion, and are associated with expansion of donor-specific Treg.


Assuntos
Diferenciação Celular/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Transplante de Células-Tronco Mesenquimais , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Condicionamento Pré-Transplante , Tolerância ao Transplante/imunologia , Animais , Células da Medula Óssea/imunologia , Feminino , Transplante de Coração/métodos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Condicionamento Pré-Transplante/métodos , Transplante Heterotópico , Transplante Homólogo , Transplante Isogênico
13.
Cell Transplant ; 29: 963689720965467, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33663249

RESUMO

Mesenchymal stromal cells (MSCs) are emerging as a novel therapeutic option for limiting chronic kidney disease progression. Conditioned medium (CM) containing bioactive compounds could convey similar benefits, avoiding the potential risks of cell therapy. This study compared the efficacy of nonrenal and renal cell-based therapy with the corresponding CM in rats with renal mass reduction (RMR). Infusions of human kidney stromal cells (kPSCs) and CM-kPSCs, but not umbilical cord (uc) MSCs or CM-ucMSCs, reduced proteinuria and preserved podocyte number and nephrin expression in RMR rats. Glomerular fibrosis, microvascular rarefaction, and apoptosis were reduced by all treatments, while the peritubular microvascular loss was reduced by kPSCs and CM-kPSCs treatment only. Importantly, kPSCs and CM-kPSCs reduced NG2-positive pericytes, and all therapies reduced α-smooth muscle actin expression, indicating reduced myofibroblast expansion. Treatment with kPSCs also significantly inhibited the accumulation of ED1-positive macrophages in the renal interstitium of RMR rats. These findings demonstrate that the CM of ucMSCs and kPSCs confers similar renoprotection as the cells. kPSCs and CM-kPSCs may be superior in attenuating chronic renal injury as a cell source.


Assuntos
Insuficiência Renal Crônica/fisiopatologia , Células Estromais/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Ratos
14.
Stem Cells ; 26(8): 2075-82, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18499895

RESUMO

Transplantation of bone marrow mesenchymal stem cells (BM-MSC) or stromal cells from rodents has been identified as a strategy for renal repair in experimental models of acute kidney injury (AKI), a highly life-threatening clinical setting. The therapeutic potential of BM-MSC of human origin has not been reported so far. Here, we investigated whether human BM-MSC treatment could prevent AKI induced by cisplatin and prolong survival in an immunodeficient mouse model. Results showed that human BM-MSC infusion decreased proximal tubular epithelial cell injury and ameliorated the deficit in renal function, resulting in reduced recipient mortality. Infused BM-MSC became localized predominantly in peritubular areas and acted to reduce renal cell apoptosis and to increase proliferation. BM-MSC also induced protection against AKI-related peritubular capillary changes consisting of endothelial cell abnormalities, leukocyte infiltration, and low endothelial cell and lumen volume density as assessed by morphometric analysis. These findings indicate that human MSC of bone marrow origin hold potential to prolong survival in AKI and should be considered for testing in a clinical trial. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células da Medula Óssea/citologia , Rim/lesões , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Animais , Apoptose , Peso Corporal , Diferenciação Celular , Proliferação de Células , Cisplatino/farmacologia , Feminino , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos SCID , Fenótipo
15.
J Am Soc Nephrol ; 18(11): 2921-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17942965

RESUMO

In mice with cisplatin-induced acute kidney injury, administration of bone marrow-derived mesenchymal stem cells (MSC) restores renal tubular structure and improves renal function, but the underlying mechanism is unclear. Here, we examined the process of kidney cell repair in co-culture experiments with MSC and cisplatin-injured proximal tubular epithelial cells (PTEC). Exposure of PTEC to cisplatin markedly reduced cell viability at 4 days, but co-culture with MSC provided a protective effect by promoting tubular cell proliferation. This effect was mediated by insulin-like growth factor-1 (IGF-1), highly expressed by MSC as mRNA and protein, since blocking the growth factor's function with a specific antibody attenuated cell proliferation of PTEC. Confirming this, knocking down IGF-1 expression in MSC by small interfering-RNA also resulted in a significant decrease in PTEC proliferation and increased apoptosis. Furthermore, in the murine model of cisplatin-induced kidney injury, administering IGF-1 gene-silenced MSC limited their protective effect on renal function and tubular structure. These findings indicate that MSC exert beneficial effects on tubular cell repair in acute kidney injury by producing the mitogenic and pro-survival factor IGF-1.


Assuntos
Células Epiteliais/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Nefropatias/terapia , Túbulos Renais Proximais/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Técnicas de Cultura de Células , Proliferação de Células , Sobrevivência Celular/fisiologia , Cisplatino , Técnicas de Cocultura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
Stem Cells Dev ; 26(17): 1258-1269, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28583028

RESUMO

Very small embryonic-like cells (VSELs) are a population of very rare pluripotent stem cells isolated in adult murine bone marrow and many other tissues and organs, including umbilical cord blood (UCB). VSEL existence is still not universally accepted by the scientific community, so for this purpose, we sought to investigate whether presumptive VSELs (pVSELs) could be isolated from human UCB with an improved protocol based on the isolation of enriched progenitor cells by depletion of nonprogenitor cells with magnetic separation. Progenitor cells, likely including VSELs, cultured with retinoic acid were able to form dense colonies and cystic embryoid bodies and to differentiate toward the ecto-meso-endoderm lineages as shown by the positivity to specific markers. VSEL differentiative potential toward mesodermal lineage was further demonstrated in vitro upon exposure to an established inductive protocol, which induced the acquisition of renal progenitor cell phenotype. VSEL-derived renal progenitors showed regenerative potential in a cisplatin model of acute kidney injury by restoring renal function and tubular structure through induction of proliferation of endogenous renal cells. The data presented here foster the great debate that surrounds VSELs and, more in general, the existence of cells endowed with pluripotent features in adult tissues. In fact, the possibility to find and isolate subpopulations of cells that fully fit all the criteria utilized to define pluripotency remains, nowadays, almost unproven. Thus, efforts to better characterize the phenotype of these intriguing cells are crucial to understand their possible applications for regenerative and precision medicine purposes.


Assuntos
Separação Celular/métodos , Sangue Fetal/citologia , Células-Tronco Pluripotentes/citologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Animais , Diferenciação Celular , Tamanho Celular , Ensaio de Unidades Formadoras de Colônias , Corpos Embrioides/citologia , Feminino , Citometria de Fluxo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Imageamento Tridimensional , Separação Imunomagnética , Rim/patologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Regeneração
17.
Biomaterials ; 27(4): 623-30, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16048730

RESUMO

Esterified hyaluronic acid (HYAFF) is routinely used for clinical tissue engineering applications such as skin and cartilage. In a previous study we developed a technique for in vitro generation of cylindrical constructs from cellularized HYAFF flat sheets. In the present investigation we studied the possibility to improve mechanical properties of this vascular construct by the addition of sodium ascorbate (SA). Non-woven HYAFF flat sheets were seeded with porcine aortic smooth muscle cells (SMCs) and cultured for 14 or 28 days with standard medium or medium added with SA. In selected experiments HYAFF sheets seeded with SMCs were wrapped to obtain cylindrical shape and then cultured in control medium or SA added medium for up to 28 days. We estimated cell viability for flat sheets, and performed histological examination, analysis of extracellular matrix (ECM) deposition and mechanical tests on tubular constructs. The number of viable cells and ECM deposition increased with time in constructs cultured in the presence of SA, as compared to control group. Moreover, SA improved mechanical properties of the vascular construct lowering material stiffness and increasing tensile strength as compared to untreated controls. The addition of SA to the medium improved cell proliferation and ECM synthesis on this biodegradable material, which leads to the formation of well organized, mechanical resistant tissue-engineered structure.


Assuntos
Ácido Ascórbico/química , Materiais Biocompatíveis/química , Prótese Vascular , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Ácido Hialurônico/química , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos/métodos , Bioprótese , Técnicas de Cultura de Células/métodos , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Elasticidade , Regeneração Tecidual Guiada/métodos , Teste de Materiais , Estresse Mecânico , Suínos , Resistência à Tração
18.
Cell Transplant ; 15 Suppl 1: S111-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16826803

RESUMO

Adult stem cells have been characterized in several tissues as a subpopulation of cells able to maintain. generate, and replace terminally differentiated cells in response to physiological cell turnover or tissue injury. Little is known regarding the presence of stem cells in the adult kidney but it is documented that under certain conditions, such as the recovery from acute injury, the kidney can regenerate itself by increasing the proliferation of some resident cells. The origin of these cells is largely undefined; they are often considered to derive from resident renal stem or progenitor cells. Whether these immature cells are a subpopulation preserved from the early stage of nephrogenesis is still a matter of investigation and represents an attractive possibility. Moreover, the contribution of bone marrow-derived stem cells to renal cell turnover and regeneration has been suggested. In mice and humans, there is evidence that extrarenal cells of bone marrow origin take part in tubular epithelium regeneration. Injury to a target organ can be sensed by bone marrow stem cells that migrate to the site of damage, undergo differentiation, and promote structural and functional repair. Recent studies have demonstrated that hematopoietic stem cells were mobilized following ischemia/reperfusion and engrafted the kidney to differentiate into tubular epithelium in the areas of damage. The evidence that mesenchymal stem cells, by virtue of their renoprotective property, restore renal tubular structure and also ameliorate renal function during experimental acute renal failure provides opportunities for therapeutic intervention.


Assuntos
Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Regeneração , Medicina Regenerativa/métodos , Células-Tronco/citologia , Animais , Terapia Genética , Humanos , Rim/citologia , Rim/fisiologia , Rim/fisiopatologia
19.
Cancer Res ; 63(7): 1534-7, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12670901

RESUMO

The tubulin-binding agent ZD6126 is a novel vascular-targeting agent in clinical development for the treatment of solid tumors. In vivo, ZD6126 is rapidly converted into N-acetylcolchinol (ZD6126 phenol). In this study, we have explored the antivascular property of N-acetylcolchinol in vitro and ZD6126 in vivo. In cell culture, N-acetylcolchinol induced rapid changes in the morphology of human umbilical vein and lung microvessel endothelial cells. Within 40 min, the compound induced endothelial cell contraction, destabilization of the tubulin cytoskeleton, induction of actin stress fibers, and membrane blebbing. These effects occurred at noncytotoxic concentrations and were rapidly reversed on removal of the drug. Nonconfluent endothelial cells were more sensitive than confluent, quiescent cells. Among different cell types, endothelial cells were the most sensitive to the induction of morphological changes, whereas smooth muscle cells were not affected. In vitro, N-acetylcolchinol rapidly disrupted a network of newly formed cords. In vivo, ZD6126 caused shut down of newly formed vessels in the Matrigel plug assay, shortly after injection. This study indicates that rapid alteration of endothelial cell morphology may be responsible for the loss of tumor blood vessel integrity, vessel shut down, and extensive tumor necrosis induced by ZD6126 in experimental tumor models.


Assuntos
Inibidores da Angiogênese/farmacologia , Colchicina/análogos & derivados , Endotélio Vascular/efeitos dos fármacos , Compostos Organofosforados/farmacologia , Animais , Células Cultivadas , Colchicina/farmacologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Pulmão/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
20.
PLoS One ; 10(3): e0120235, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25811887

RESUMO

New intervention tools for severely damaged kidneys are in great demand to provide patients with a valid alternative to whole organ replacement. For repairing or replacing injured tissues, emerging approaches focus on using stem and progenitor cells. Embryonic kidneys represent an interesting option because, when transplanted to sites such as the renal capsule of healthy animals, they originate new renal structures. Here, we studied whether metanephroi possess developmental capacity when transplanted under the kidney capsule of MWF male rats, a model of spontaneous nephropathy. We found that six weeks post-transplantation, renal primordia developed glomeruli and tubuli able to filter blood and to produce urine in cyst-like structures. Newly developed metanephroi were able to initiate a regenerative-like process in host renal tissues adjacent to the graft in MWF male rats as indicated by an increase in cell proliferation and vascular density, accompanied by mRNA and protein upregulation of VEGF, FGF2, HGF, IGF-1 and Pax-2. The expression of SMP30 and NCAM was induced in tubular cells. Oxidative stress and apoptosis markedly decreased. Our study shows that embryonic kidneys generate functional nephrons when transplanted into animals with severe renal disease and at the same time activate events at least partly mimicking those observed in kidney tissues during renal regeneration.


Assuntos
Nefropatias/terapia , Transplante de Rim , Regeneração , Animais , Apoptose , Biomarcadores , Proliferação de Células , Feminino , Fibroblastos/metabolismo , Fibroblastos/transplante , Expressão Gênica , Sobrevivência de Enxerto , Nefropatias/patologia , Nefropatias/fisiopatologia , Transplante de Rim/métodos , Masculino , Neovascularização Fisiológica , Estresse Oxidativo , Ratos , Regeneração/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa