Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alvaro Pacheco-Silva is active.

Publication


Featured researches published by Alvaro Pacheco-Silva.


International Immunopharmacology | 2009

Early modulation of inflammation by mesenchymal stem cell after acute kidney injury

Patricia Semedo; Carolina G. Palasio; Cassiano D. Oliveira; Carla Q. Feitoza; Giselle Martins Gonçalves; Marcos Antonio Cenedeze; Pamella M.H. Wang; Vicente de Paula Antunes Teixeira; Marlene Antônia dos Reis; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Therapy with stem cells has showed to be promising for acute kidney injury (AKI), although how it works is still controversial. Modulation of the inflammatory response is one possible mechanism. Most of published data relies on early time and whether the protection is still maintained after that is not known. Here, we analyzed whether immune modulation continues after 24 h of reperfusion. MSC were obtained from male Wistar rats. After 3-5 passages, cells were screened for CD73, CD90, CD44, CD45, CD29 and CD 31. In addition, MSC were submitted to differentiation in adipocyte and in osteocyte. AKI was induced by bilaterally clamping of renal pedicles for 60 min. Six hours after injury, MSC (2 x 10(5) cells) were administered intravenously. MSC-treated animals presented the lowest serum creatinine compared to non-treated animals (24 h: 1.3+/-0.21 vs. 3.23+/-0.89 mg/dl, p<0.05). The improvement in renal function was followed by a lower expression of IL-1b, IL-6 and TNF-alpha and higher expression of IL-4 and IL-10. However, 48 h after reperfusion, this cytokine profile has changed. The decrease in Th1 cytokines was less evident and IL-6 was markedly up regulated. PCNA analysis showed that regeneration occurs faster in kidney tissues of MSC-treated animals than in controls at 24 h. And also ratio of Bcl-2/Bad was higher at treated animals after 24 and 48 h. Our data demonstrated that the immunomodulatory effects of MSC occur at very early time point, changing the inflammation profile toward a Th2 profile.


Diabetes | 2012

Immune Regulatory Properties of Allogeneic Adipose-Derived Mesenchymal Stem Cells in the Treatment of Experimental Autoimmune Diabetes

Ênio José Bassi; Pedro M. Moraes-Vieira; Carla S.R. Moreira-Sá; Danilo Candido de Almeida; Leonardo M. Vieira; Cláudia da Silva Cunha; Meire Ioshie Hiyane; Alexandre S. Basso; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Adipose-derived mesenchymal stem cells (ADMSCs) display immunosuppressive properties, suggesting a promising therapeutic application in several autoimmune diseases, but their role in type 1 diabetes (T1D) remains largely unexplored. The aim of this study was to investigate the immune regulatory properties of allogeneic ADMSC therapy in T cell–mediated autoimmune diabetes in NOD mice. ADMSC treatment reversed the hyperglycemia of early-onset diabetes in 78% of diabetic NOD mice, and this effect was associated with higher serum insulin, amylin, and glucagon-like peptide 1 levels compared with untreated controls. This improved outcome was associated with downregulation of the CD4+ Th1-biased immune response and expansion of regulatory T cells (Tregs) in the pancreatic lymph nodes. Within the pancreas, inflammatory cell infiltration and interferon-γ levels were reduced, while insulin, pancreatic duodenal homeobox-1, and active transforming growth factor-β1 expression were increased. In vitro, ADMSCs induced the expansion/proliferation of Tregs in a cell contact–dependent manner mediated by programmed death ligand 1. In summary, ADMSC therapy efficiently ameliorates autoimmune diabetes pathogenesis in diabetic NOD mice by attenuating the Th1 immune response concomitant with the expansion/proliferation of Tregs, thereby contributing to the maintenance of functional β-cells. Thus, this study may provide a new perspective for the development of ADMSC-based cellular therapies for T1D.


American Journal of Transplantation | 2005

Early presence of calcium oxalate deposition in kidney graft biopsies is associated with poor long-term graft survival

Hélady Sanders Pinheiro; Niels Olsen Saraiva Camara; Kikumi Suzete Osaki; Luiz Antonio Ribeiro de Moura; Alvaro Pacheco-Silva

Accumulated oxalate will be excreted after renal transplantation, creating an increased risk of tubular precipitation, especially in the presence of allograft dysfunction. We evaluated calcium oxalate (CaOx) deposition in renal allograft biopsies with early dysfunction, its association with acute tubular necrosis (ATN) and graft survival. We studied 97 renal transplant patients, submitted to a graft biopsy within 3 months post‐transplant, and reanalyzed them after 10 years. We analyzed renal tissue under polarized light and quantified CaOx deposits. CaOx deposits were detected in 52.6% of the patients; 26.8% were of mild and 25.8% of moderate intensity. The deposits were more frequent in biopsies performed within 3 weeks post‐transplant (82.4 vs. 63.0%, p < 0.05) and in allografts with more severe renal dysfunction (creatinine 5.6 mg/dL vs. 3.4 mg/dL, p < 0.001). ATN incidence was also higher in patients with CaOx deposits (47% vs. 24%, p < 0.001). Twelve‐year graft survival was strikingly worse in patients with CaOx deposits compared to those free of deposits (49.7 vs. 74.1%, p = 0.013). Our study shows a high incidence of CaOx deposits in kidney allografts with early dysfunction, implying an additional risk for acute tubular injury, with a negative impact on graft survival.


PLOS ONE | 2012

TLR2, TLR4 and the MYD88 Signaling Pathway Are Crucial for Neutrophil Migration in Acute Kidney Injury Induced by Sepsis

Angela Castoldi; Tarcio Teodoro Braga; Matheus Correa-Costa; Cristhiane Fávero Aguiar; Ênio José Bassi; Reinaldo Correa-Silva; Rosa Maria Elias; Fábia Andréia Salvador; Pedro M. Moraes-Vieira; Marcos Antonio Cenedeze; Marlene Antônia dos Reis; Meire Ioshie Hiyane; Alvaro Pacheco-Silva; Giselle Martins Gonçalves; Niels Olsen Saraiva Câmara

The aim of this study was to investigate the role of TLR2, TLR4 and MyD88 in sepsis-induced AKI. C57BL/6 TLR2−/−, TLR4−/− and MyD88−/− male mice were subjected to sepsis by cecal ligation and puncture (CLP). Twenty four hours later, kidney tissue and blood samples were collected for analysis. The TLR2−/−, TLR4−/− and MyD88−/− mice that were subjected to CLP had preserved renal morphology, and fewer areas of hypoxia and apoptosis compared with the wild-type C57BL/6 mice (WT). MyD88−/− mice were completely protected compared with the WT mice. We also observed reduced expression of proinflammatory cytokines in the kidneys of the knockout mice compared with those of the WT mice and subsequent inhibition of increased vascular permeability in the kidneys of the knockout mice. The WT mice had increased GR1+low cells migration compared with the knockout mice and decreased in GR1+high cells migration into the peritoneal cavity. The TLR2−/−, TLR4−/−, and MyD88−/− mice had lower neutrophil infiltration in the kidneys. Depletion of neutrophils in the WT mice led to protection of renal function and less inflammation in the kidneys of these mice. Innate immunity participates in polymicrobial sepsis-induced AKI, mainly through the MyD88 pathway, by leading to an increased migration of neutrophils to the kidney, increased production of proinflammatory cytokines, vascular permeability, hypoxia and apoptosis of tubular cells.


Clinical Transplantation | 2007

The use of sirolimus in ganciclovir-resistant cytomegalovirus infections in renal transplant recipients.

Kikumi Suzete Ozaki; Niels Olsen Saraiva Câmara; Eliana Nogueira; Mauricio Galvão Pereira; Celso Francisco Hernandes Granato; Cláudio Melaragno; Luis Fernando Aranha Camargo; Alvaro Pacheco-Silva

Abstract:  Background:  The widespread use of prophylactic ganciclovir and anti‐lymphocyte/thymocyte therapies are associated with increased induction of ganciclovir‐resistant cytomegalovirus (CMV) strains. The use of sirolimus has been associated with a lower incidence of CMV infection in transplant recipients. We questioned whether it could also be effective as a therapeutic treatment of resistant CMV infection.


Molecular Medicine | 2012

MyD88 signaling pathway is involved in renal fibrosis by favoring a TH2 immune response and activating alternative M2 macrophages.

Tarcio Teodoro Braga; Matheus Correa-Costa; Yuri Felipe Souza Guise; Angela Castoldi; Cassiano D. Oliveira; Meire Ioshie Hyane; Marcos Antonio Cenedeze; Simone A. Teixeira; Marcelo N. Muscará; Katia R. Perez; Iolanda M. Cuccovia; Alvaro Pacheco-Silva; Giselle Martins Gonçalves; Niels Olsen Saraiva Camara

Inflammation contributes to the pathogenesis of chronic kidney disease (CKD). Molecules released by the inflamed injured tissue can activate toll-like receptors (TLRs), thereby modulating macrophage and CD4+ T-cell activity. We propose that in renal fibrogenesis, M2 macrophages are recruited and activated in a T helper subset 2 cell (TH2)-prone inflammatory milieu in a MyD88-dependent manner. Mice submitted to unilateral ureteral ligation (UUO) demonstrated an increase in macrophage infiltration with collagen deposition after 7 d. Conversely, TLR2, TLR4 and MyD88 knockout (KO) mice had an improved renal function together with diminished TH2 cytokine production and decreased fibrosis formation. Moreover, TLR2, TLR4 and MyD88 KO animals exhibited less M2 macrophage infiltration, namely interleukin (IL)-10+ and CD206+ CDllbhigh cells, at 7 d after surgery. We evaluated the role of a TH2 cytokine in this context, and observed that the absence of IL-4 was associated with better renal function, decreased IL-13 and TGF-β levels, reduced arginase activity and a decrease in fibrosis formation when compared with IL-12 KO and wild-type (WT) animals. Indeed, the better renal outcomes and the decreased fibrosis formation were restricted to the deficiency of IL-4 in the hematopoietic compartment. Finally, macrophage depletion, rather than the absence of T cells, led to reduced lesions of the glomerular filtration barrier and decreased collagen deposition. These results provide evidence that future therapeutic strategies against renal fibrosis should be accompanied by the modulation of the M1:M2 and TH1:TH2 balance, as TH2 and M2 cells are predictive of fibrosis toward mechanisms that are sensed by innate immune response and triggered in a MyD88-dependent pathway.


PLOS ONE | 2010

Induction of Heme Oxygenase-1 Can Halt and Even Reverse Renal Tubule-Interstitial Fibrosis

Matheus Correa-Costa; Patricia Semedo; Ana Paula Fernandes da Silva Monteiro; Reinaldo Correia Silva; Rafael Luiz Pereira; Giselle Martins Gonçalves; Geórgia D.M. Marques; Marcos Antonio Cenedeze; Ana Carolina Guimarães Faleiros; Alexandre C. Keller; Maria Heloisa Massola Shimizu; Antonio Carlos Seguro; Marlene Antônia dos Reis; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Background The tubule-interstitial fibrosis is the hallmark of progressive renal disease and is strongly associated with inflammation of this compartment. Heme-oxygenase-1 (HO-1) is a cytoprotective molecule that has been shown to be beneficial in various models of renal injury. However, the role of HO-1 in reversing an established renal scar has not yet been addressed. Aim We explored the ability of HO-1 to halt and reverse the establishment of fibrosis in an experimental model of chronic renal disease. Methods Sprague-Dawley male rats were subjected to unilateral ureteral obstruction (UUO) and divided into two groups: non-treated and Hemin-treated. To study the prevention of fibrosis, animals were pre-treated with Hemin at days -2 and -1 prior to UUO. To investigate whether HO-1 could reverse established fibrosis, Hemin therapy was given at days 6 and 7 post-surgery. After 7 and/or 14 days, animals were sacrificed and blood, urine and kidney tissue samples were collected for analyses. Renal function was determined by assessing the serum creatinine, inulin clearance, proteinuria/creatininuria ratio and extent of albuminuria. Arterial blood pressure was measured and fibrosis was quantified by Picrosirius staining. Gene and protein expression of pro-inflammatory and pro-fibrotic molecules, as well as HO-1 were performed. Results Pre-treatment with Hemin upregulated HO-1 expression and significantly reduced proteinuria, albuminuria, inflammation and pro-fibrotic protein and gene expressions in animals subjected to UUO. Interestingly, the delayed treatment with Hemin was also able to reduce renal dysfunction and to decrease the expression of pro-inflammatory molecules, all in association with significantly reduced levels of fibrosis-related molecules and collagen deposition. Finally, TGF-β protein production was significantly lower in Hemin-treated animals. Conclusion Treatment with Hemin was able both to prevent the progression of fibrosis and to reverse an established renal scar. Modulation of inflammation appears to be the major mechanism behind HO-1 cytoprotection.


Cell Transplantation | 2012

Adipose tissue-derived stem cell treatment prevents renal disease progression.

Cassiano Donizetti-Oliveira; Patricia Semedo; Marina Burgos-Silva; Marco Antonio Cenedeze; Denise Maria Avancini Costa Malheiros; Marlene Antônia dos Reis; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Adipose tissue-derived stem cells (ASCs) are an attractive source of stem cells with regenerative properties that are similar to those of bone marrow stem cells. Here, we analyze the role of ASCs in reducing the progression of kidney fibrosis. Progressive renal fibrosis was achieved by unilateral clamping of the renal pedicle in mice for 1 h; after that, the kidney was reperfused immediately. Four hours after the surgery, 2 × 105 ASCs were intraperitoneally administered, and mice were followed for 24 h posttreatment and then at some other time interval for the next 6 weeks. Also, animals were treated with 2 × 105 ASCs at 6 weeks after reperfusion and sacrificed 4 weeks later to study their effect when interstitial fibrosis is already present. At 24 h after reperfusion, ASC-treated animals showed reduced renal dysfunction and enhanced regenerative tubular processes. Renal mRNA expression of IL-6 and TNF was decreased in ASC-treated animals, whereas IL-4, IL-10, and HO-1 expression increased despite a lack of ASCs in the kidneys as determined by SRY analysis. As expected, untreated kidneys shrank at 6 weeks, whereas the kidneys of ASC-treated animals remained normal in size, showed less collagen deposition, and decreased staining for FSP-1, type I collagen, and Hypoxyprobe. The renal protection seen in ASC-treated animals was followed by reduced serum levels of TNF-α, KC, RANTES, and IL-1α. Surprisingly, treatment with ASCs at 6 weeks, when animals already showed installed fibrosis, demonstrated amelioration of functional parameters, with less tissue fibrosis observed and reduced mRNA expression of type I collagen and vimentin. ASC therapy can improve functional parameters and reduce progression of renal fibrosis at early and later times after injury, mostly due to early modulation of the inflammatory response and to less hypoxia, thereby reducing the epithelial–mesenchymal transition.


Transplant International | 2008

A Role for galectin‐3 in renal tissue damage triggered by ischemia and reperfusion injury

A.P.F. Bertocchi; Gabriela Campanhole; Pamella Huey Mei Wang; Giselle Martins Gonçalves; M.J. Damião; Marcos Antonio Cenedeze; Felipe Caetano Beraldo; Vicente de Paula Antunes Teixeira; Marlene Antônia dos Reis; Marilda Mazzali; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Ischemic‐reperfusion injury (IRI) triggers an inflammatory response involving neutrophils/macrophages, lymphocytes and endothelial cells. Galectin‐3 is a multi‐functional lectin with a broad range of action such as promotion of neutrophil adhesion, induction of oxidative stress, mastocyte migration and degranulation, and production of pro‐inflammatory cytokines. The aim of this study was evaluate the role of galectin‐3 in the inflammation triggered by IRI. Galectin‐3 knockout (KO) and wild type (wt) mice were subjected to 45 min of renal pedicle occlusion. Blood and kidney samples were collected at 6, 24, 48 and 120 h. Blood urea was analyzed enzymatically, while MCP‐1, IL‐6 and IL‐1β were studied by real‐time PCR. Reactive oxygen species (ROS) was investigated by flow cytometry. Morphometric analyses were performed at 6, 24, 48 and 120 h after reperfusion. Urea peaked at 24 h, being significantly lower in knockout animals (wt = 264.4 ± 85.21 mg/dl vs. gal‐3 KO = 123.74 ± 29.64 mg/dl, P = 0.001). Galectin‐3 knockout animals presented less acute tubular necrosis and a more prominent tubular regeneration when compared with controls concurrently with lower expression of MCP‐1, IL‐6, IL‐1β, less macrophage infiltration and lower ROS production at early time points. Galectin‐3 seems to play a role in renal IRI involving the secretion of macrophage‐related chemokine, pro‐inflammatory cytokines and ROS production.


American Journal of Transplantation | 2007

High Expression of Tim-3 mRNA in Urinary Cells from Kidney Transplant Recipients with Acute Rejection

Paulo Guilherme Renesto; Viviane Campos Ponciano; Marcos Antonio Cenedeze; N.O. Saraiva Câmara; Alvaro Pacheco-Silva

TIM‐3 is a recently described molecule specifically expressed on Th1 differentiated T cells. We explored the usefulness of urinary mRNA profiles in the diagnosis of renal acute rejection (AR). Sixty urinary samples from renal transplant recipients simultaneously collected to allograft biopsy, (AR = 30 and No‐AR =30), and 12 urinary samples from stable renal transplants were analyzed. Urinary mRNA encoding for TIM‐3 and IFN‐γ were quantified using real time RT‐PCR. TIM‐3 mRNA was highly expressed in AR (559.19 ± 644.41) compared to No‐AR (3.78 ± 7.20), and stable transplants (0.54 ± 0.76), p < 0.001. To a lesser degree, IFN‐γ mRNA transcripts were also increased in AR (50.40 ± 38.71), compared with No‐AR (4.69 ± 12.62), and stable transplants (0.38 ± 0.44) p < 0.001. The highest expression of TIM‐3 in AR makes it a promising noninvasive test for its diagnosis.

Collaboration


Dive into the Alvaro Pacheco-Silva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcos Antonio Cenedeze

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Giselle Martins Gonçalves

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Patricia Semedo

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K.S. Ozaki

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Niels Olsen Saraiva Camara

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge