Network


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

Hotspot


Dive into the research topics where Ana J. Coito is active.

Publication


Featured researches published by Ana J. Coito.


Journal of Clinical Investigation | 1999

Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury

Farin Amersi; Roland Buelow; Hirohisa Kato; Bibo Ke; Ana J. Coito; Xiu Da Shen; Delai Zhao; Joseph Zaky; Judy Melinek; Charles Lassman; Jay K. Kolls; Jawed Alam; Thomas Ritter; H.-D. Volk; Douglas G. Farmer; Rafik M. Ghobrial; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

We examined the effects of upregulation of heme oxygenase-1 (HO-1) in steatotic rat liver models of ex vivo cold ischemia/reperfusion (I/R) injury. In the model of ischemia/isolated perfusion, treatment of genetically obese Zucker rats with the HO-1 inducer cobalt protoporphyrin (CoPP) or with adenoviral HO-1 (Ad-HO-1) significantly improved portal venous blood flow, increased bile production, and decreased hepatocyte injury. Unlike in untreated rats or those pretreated with the HO-1 inhibitor zinc protoporphyrin (ZnPP), upregulation of HO-1 by Western blots correlated with amelioration of histologic features of I/R injury. Adjunctive infusion of ZnPP abrogated the beneficial effects of Ad-HO-1 gene transfer, documenting the direct involvement of HO-1 in protection against I/R injury. Following cold ischemia/isotransplantation, HO-1 overexpression extended animal survival from 40% in untreated controls to about 80% after CoPP or Ad-HO-1 therapy. This effect correlated with preserved hepatic architecture, improved liver function, and depressed infiltration by T cells and macrophages. Hence, CoPP- or gene therapy-induced HO-1 prevented I/R injury in steatotic rat livers. These findings provide the rationale for refined new treatments that should increase the supply of usable donor livers and ultimately improve the overall success of liver transplantation.


American Journal of Transplantation | 2001

Heme oxygenase-1 overexpression protects rat livers from ischemia/reperfusion injury with extended cold preservation.

Hirohisa Kato; Farin Amersi; Roland Buelow; Judy Melinek; Ana J. Coito; Bibo Ke; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

This study analyzes the effects and mechanisms of heme oxygenase‐1 (HO‐1)‐mediated cytoprotection in rat livers exposed to cold preservation. In the first series, rats were pretreated with cobalt protoporphyrin (CoPP) or zinc protoporphyrin (ZnPP), HO‐1 inducer and antagonist, respectively. Livers were stored at 4°C for 24 h, and then perfused ex vivo for 2 h. Livers pretreated with CoPP had significantly higher portal venous blood flow and increased total bile production, as compared with the ZnPP group. This correlated with histologic (Banff) criteria of hepatocyte injury/liver function. In the second series, rat livers were stored at 4 °C for 24 h or 40 h, and then transplanted into syngeneic recipients. After 24 h of preservation, 80% of rats bearing CoPP‐pretreated liver grafts survived 21 days (vs. 50% in controls). After 40 h of cold preservation, liver transplant survival at day 1, 7 and 21 for the CoPP group was: 100%, 71% and 57%, respectively (vs. 50%, 50% and 33% in controls). This correlated with improved hepatic function/histologic (Suzuki) criteria of hepatocyte injury after HO‐1 overexpression (immunohistology/Western blots) by infiltrating macrophages. This study documents the potential utility of HO‐1‐inducing agents in preventing ischemia/reperfusion injury resulting from prolonged storage of liver transplants.


Transplantation | 2002

Heme oxygenase-1 overexpression protects rat hearts from cold ischemia/reperfusion injury via an antiapoptotic pathway.

Masamichi Katori; Roland Buelow; Bibo Ke; Jeff Ma; Ana J. Coito; Suhasini Iyer; Daniel Southard; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

BACKGROUND Ischemia/reperfusion (I/R) injury is one of the most important causes of the early graft loss. We have shown that overexpression of heme oxygenase-1 (HO-1), an inducible heat shock protein 32, protects rat livers against I/R injury. We report on the cytoprotective effects of HO-1 in a rat cardiac I/R injury model, using cobalt protoporphyrin (CoPP) as HO-1 inducer and zinc protoporphyrin (ZnPP) as HO-1 inhibitor. METHODS Three groups of Lewis rats were studied: group 1 control donors received phosphate-buffered saline 48 hr before the harvest; group 2 donors were pretreated with CoPP at -48 hr; and in group 3, donors received CoPP at -48 hr and ZnPP was given to recipients at reperfusion. Hearts were harvested, stored in University of Wisconsin solution (4 degrees C) for 24 hr, and then transplanted to syngeneic (Lewis) rats. RESULTS Sixty percent of control grafts ceased their function in <15 min. In contrast, 80% of CoPP-pretreated grafts survived 14 days. All grafts stopped functioning within 24 hr after CoPP + ZnPP therapy. Cardiac HO-1 enzymatic activity and protein expression correlated with beneficial effects of CoPP and deleterious effects of adjunctive ZnPP treatment. Markedly less apoptotic (TUNEL+) myocyte/endothelial cells could be detected in CoPP cardiac grafts, as compared with controls. The expression of antiapoptotic (Bcl-2/Bag-1) proteins was up-regulated in the CoPP group. CONCLUSION HO-1 overexpression provides potent protection against cold I/R injury in a stringent rat cardiac model. This effect depends, at least in part, on HO-1-mediated up-regulation of a host antiapoptotic mechanism, especially in the early postreperfusion period.


Transplantation | 2002

Heme oxygenase-1 gene transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia-reperfusion injury.

Ana J. Coito; Roland Buelow; Xiu-Da Shen; Farin Amersi; Carolina Moore; Hans-Dieter Volk; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

BACKGROUND Ischemia/reperfusion (I/R) injury is a critical factor in the dysfunction of steatotic orthotopic liver transplants. Heme oxygenase-1 (HO-1), a cytoprotective protein, may be important in ameliorating hepatic I/R injury. METHODS We used adenovirus (Ad)-based HO-1 gene transfer to analyze the effects of HO-1 overexpression in a well-established fatty Zucker rat model of I/R followed by orthotopic liver transplantation. RESULTS Ad-HO-1 gene therapy increased recipient survival (80% vs. 40-50% in controls) and significantly diminished hepatocyte injury, as compared with untreated and Ad-beta-galactosidase (Ad-beta-Gal)-treated livers. Orthotopic liver transplants in the Ad-HO-1 group exhibited less macrophage infiltration in the portal areas, as compared with controls. Unlike untreated and Ad-beta-Gal-treated orthotopic liver transplant controls, which showed elevated levels of inducible nitric oxide synthase by infiltrating macrophages, inducible nitric oxide synthase expression in the Ad-HO-1 group was almost absent. In contrast, endothelial nitric oxide synthase was comparable in Ad-HO-1- and Ad-beta-Gal-transduced fatty orthotopic liver transplants. Intragraft expression of antiapoptotic Bcl-2 and Bag-1 was increased in Ad-HO-1-treated orthotopic liver transplants, as compared with Ad-beta-Gal controls. Moreover, increased HO enzymatic activity was accompanied by inhibition of caspase-3 protein expression. CONCLUSIONS HO-1 gene transfer significantly prolongs survival of steatotic orthotopic liver transplants, depresses macrophage infiltration, suppresses local expression of inducible nitric oxide synthase, and modulates pro- and antiapoptotic pathways.


Journal of The American Society of Nephrology | 2003

Gene Transfer-Induced Local Heme Oxygenase-1 Overexpression Protects Rat Kidney Transplants From Ischemia/Reperfusion Injury

Tom Blydt-Hansen; Masamichi Katori; Charles Lassman; Bibo Ke; Ana J. Coito; Suhasani Iyer; Roland Buelow; Robert B. Ettenger; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

Heme oxygenase-1 (HO-1) overexpression using gene transfer protects rat livers against ischemia/reperfusion (I/R) injury. This study evaluates the effects of Ad-HO-1 gene transfer in a rat renal isograft model. Donor LEW kidneys were perfused with Ad-HO-1, Ad-beta-gal, or PBS, stored at 4 degrees C for 24 h, and transplanted orthotopically into LEW recipients, followed by contralateral native nephrectomy. Serum creatinine, urine protein/creatinine ratios, severity of histologic changes, HO-1 mRNA/protein expression, and HO enzymatic activity were analyzed. Ad-HO-1 gene transfer conferred a survival advantage when compared with PBS- and Ad-beta-gal-treated controls, with median survival of 100, 7, and 7 d, respectively (P < 0.01). Serum creatinine levels were elevated at day 7 in all groups (range, 2.2 to 5.8 mg/dl) but recovered to 1.0 mg/dl by day 14 (P < 0.01) in Ad-HO-1 group, which was sustained thereafter. Urine protein/creatinine ratio at day 7 was elevated in both PBS and Ad-beta-gal, as compared with the Ad-HO-1 group (12.0 and 9.8 versus 5.0; P < 0.005); histologically, ATN and glomerulosclerosis was more severe in Ad-beta-gal group at all time points. Reverse transcriptase-PCR-based HO-1 gene expression was significantly increased before reperfusion (P < 0.001) and remained increased in the Ad-HO-1-treated group for 3 d after transplantation. Concomitantly, HO enzymatic activity was increased at transplantation and at 3 d posttransplant in the Ad-HO-1 group, compared with Ad-beta-gal controls (P < 0.05); tubular HO-1 expression was discernible early posttransplant in the Ad-HO-1 group alone. These findings are consistent with protective effects of HO-1 overexpression using a gene transfer approach against severe renal I/R injury, with reduced mortality and attenuation of tissue injury.


Hepatology | 2007

Metalloproteinase‐9 deficiency protects against hepatic ischemia/reperfusion injury

Takashi Hamada; Constantino Fondevila; Ronald W. Busuttil; Ana J. Coito

Leukocyte transmigration across endothelial and extracellular matrix protein barriers is dependent on adhesion and focal matrix degradation events. In the present study we investigated the role of metalloproteinase‐9 (MMP‐9/gelatinase B) in liver ischemia/reperfusion (I/R) injury using MMP‐9‐deficient (MMP‐9−/−) animals and mice treated with a specific anti‐MMP‐9 neutralizing antibody or with a broad gelatinase inhibitor for both MMP‐9 and metalloproteinase‐2 (MMP‐2/gelatinase A). Compared to wild‐type mice, MMP‐9−/− mice and mice treated with an anti‐MMP‐9 antibody showed significantly reduced liver damage. In contrast, mice treated with a broad gelatinase inhibitor showed rather inferior protection against I/R injury and were characterized by persistent ongoing liver inflammation, suggesting that MMP‐2 and MMP‐9 may have distinct roles in this type of injury. MMP‐9 was mostly detected in Ly‐6G and macrophage antigen–1 leukocytes adherent to the vessel walls and infiltrating the damaged livers of wild‐type mice after liver I/R injury. Leukocyte traffic and cytokine expression were markedly impaired in livers of MMP‐9−/− animals and in livers of mice treated with anti‐MMP‐9 antibody after I/R injury; however, initiation of the endothelial adhesion cascades was similar in both MMP‐9−/− and control livers. We also showed that MMP‐9‐specific inhibition disrupted neutrophil migration across fibronectin in transwell filters and depressed myeloperoxidase (MPO) activation in vitro. Conclusion: These results support critical functions for MMP‐9 in leukocyte recruitment and activation leading to liver damage. Moreover, they provide the rationale for identifying inhibitors to specifically target MMP‐9 in vivo as a potential therapeutic approach in liver I/R injury. (HEPATOLOGY 2007.)


Matrix Biology | 2015

Matrix metalloproteinases in liver injury, repair and fibrosis

Sergio Duarte; John T. Baber; Takehiro Fujii; Ana J. Coito

The liver is a large highly vascularized organ with a central function in metabolic homeostasis, detoxification, and immunity. Due to its roles, the liver is frequently exposed to various insults which can cause cell death and hepatic dysfunction. Alternatively, the liver has a remarkable ability to self-repair and regenerate after injury. Liver injury and regeneration have both been linked to complex extracellular matrix (ECM) related pathways. While normal degradation of ECM components is an important feature of tissue repair and remodeling, irregular ECM turnover contributes to a variety of liver diseases. Matrix metalloproteinases (MMPs) are the main enzymes implicated in ECM degradation. MMPs not only remodel the ECM, but also regulate immune responses. In this review, we highlight some of the MMP-attributed roles in acute and chronic liver injury and emphasize the need for further experimentation to better understand their functions during hepatic physiological conditions and disease progression.


American Journal of Transplantation | 2002

FTY720 Pretreatment Reduces Warm Hepatic Ischemia Reperfusion Injury Through Inhibition of T‐Lymphocyte Infiltration

Dean M. Anselmo; Farin Amersi; Xiu-Da Shen; Feng Gao; Masamichi Katori; Charles Lassman; Bibo Ke; Ana J. Coito; Jeffrey Ma; Volker Brinkmann; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski; Douglas G. Farmer

Ischemia and reperfusion (IR) injury remains a significant problem in clinical liver transplantation. We investigated the effects of lymphocyte depletion with FTY720 in models of warm hepatic IR. Using 60‐min partial warm hepatic IR, three groups of rats were studied: Sham – laparotomy alone; Control – water p.o. × 3 d before ischemia; Treatment – FTY720 p.o. × 3 d before ischemia. Animals were sacrificed for analysis at 6 h and 24 h post reperfusion. The effect of FTY720 pretreatment on survival was also studied using 150 min total hepatic IR with portojugular shunt. FTY720 treatment significantly reduced serum glutamic pyruvic transaminase and peripheral blood lymphocytes compared to controls at 6 h and 24 h (p < 0.0005). Histological grade was significantly improved in treated livers vs. controls (p < 0.05). CD3 immunocytochemical analysis revealed a significant reduction in T‐cell infiltration in FTY720‐treated livers (p < 0.0002). No difference in tissue myeloperoxidase levels was observed. Seven‐day survival was significantly improved in treated rats vs. controls following total hepatic ischemia (p < 0.05). In conclusion, FTY720 ameliorates the biochemical and histological manifestations of hepatic IR by preventing T‐lymphocyte infiltration and prolongs survival following a more severe ischemic insult. Myeloperoxidase data suggest this mechanism is independent of neutrophil activation. These results indicate that T lymphocytes are pivotal mediators in hepatic IR and may have important implications in liver transplantation.


American Journal of Transplantation | 2002

P‐Selectin Glycoprotein Ligand‐1 (rPSGL‐Ig)‐Mediated Blockade of CD62 Selectin Molecules Protects Rat Steatotic Liver Grafts from Ischemia/Reperfusion Injury

Farin Amersi; Douglas G. Farmer; Gray D. Shaw; Hirohisa Kato; Ana J. Coito; Fady M. Kaldas; Delai Zhao; Charles Lassman; Judy Melinek; Jeffrey Ma; Hans-Dieter Volk; Jerzy W. Kupiec-Weglinski; Ronald W. Busuttil

We examined the effects of early blockade of CD62 selectin‐mediated adhesive interactions in steatotic rat liver models of ex vivo cold ischemia followed by reperfusion or transplantation by administration of P‐selectin glycoprotein ligand‐1 (rPSGL‐Ig). In the model of cold ischemia/reperfusion, livers pretreated ex vivo with rPSGL‐Ig at harvesting from obese Zucker rats showed significantly decreased portal resistance, increased bile production, and diminished hepatic endothelial neutrophil infiltration, as compared with untreated controls. Pretreatment of fatty livers with rPSGL‐Ig prior to transplantation extended the survival of lean Zucker rat recipients from 40% to 90%. This effect correlated with significantly improved liver function, depressed neutrophil activity, and decreased histologic features of hepatocyte injury. Intragraft expression of CD62 P‐selectin was similar in both recipient groups. rPSGL‐Ig treatment decreased intragraft infiltration by CD3/CD25 cells, diminished expression of pro‐inflammatory TNFα, IL‐6, iNOS, IL‐2 and IFN‐γ, without significantly affecting mRNA levels coding for anti‐inflammatory IL‐4. Thus, rPSGL‐Ig blockade of CD62‐mediated adhesive interactions protects against severe ischemia/reperfusion injury suffered otherwise by steatotic rat livers. These findings document the potential utility of rPSGL‐Ig in increasing the transplant donor pool through modulation of marginal steatotic livers.


Journal of Immunology | 2008

Cyclooxygenase-2 Deficiency Enhances Th2 Immune Responses and Impairs Neutrophil Recruitment in Hepatic Ischemia/Reperfusion Injury

Takashi Hamada; Sei-ichiro Tsuchihashi; Armine Avanesyan; Sergio Duarte; Carolina Moore; Ronald W. Busuttil; Ana J. Coito

Cyclooxygenase-2 (COX-2) is a prostanoid-synthesizing enzyme that is critically implicated in a variety of pathophysiological processes. Using a COX-2-deficient mouse model, we present data that suggest that COX-2 has an active role in liver ischemia/reperfusion (I/R) injury. We demonstrate that COX-2-deficient mice had a significant reduction in liver damage after I/R insult. The inability of COX-2−/− to elaborate COX-2 products favored a Th2-type response in these mice. COX-2−/− livers after I/R injury showed significantly decreased levels of IL-2, as well as IL-12, a cytokine known to have a central role in Th1 effector cell differentiation. Moreover, such livers expressed enhanced levels of the anti-inflammatory cytokine IL-10, shifting the balance in favor of a Th2 response in COX-2-deficient mice. The lack of COX-2 expression resulted in decreased levels of CXCL2, a neutrophil-activating chemokine, reduced infiltration of MMP-9-positive neutrophils, and impaired late macrophage activation in livers after I/R injury. Additionally, Bcl-2 and Bcl-xL were normally expressed in COX-2−/− livers after injury, whereas respective wild-type controls were almost depleted of these two inhibitors of cell death. In contrast, caspase-3 activation and TUNEL-positive cells were depressed in COX-2−/− livers. Therefore, our data support the concept that COX-2 is involved in the pathogenic events occurring in liver I/R injury. The data also suggest that potential valuable therapeutic approaches in liver I/R injury may result from further studies aimed at identifying specific COX-2-derived prostanoid pathways.

Collaboration


Dive into the Ana J. Coito's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiu-Da Shen

University of California

View shared research outputs
Top Co-Authors

Avatar

Sergio Duarte

University of California

View shared research outputs
Top Co-Authors

Avatar

Bibo Ke

University of California

View shared research outputs
Top Co-Authors

Avatar

Feng Gao

University of California

View shared research outputs
Top Co-Authors

Avatar

Carolina Moore

University of California

View shared research outputs
Top Co-Authors

Avatar

Farin Amersi

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takashi Hamada

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge