Network


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

Hotspot


Dive into the research topics where Masamichi Katori is active.

Publication


Featured researches published by Masamichi Katori.


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.


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.


Transplant Immunology | 2002

A novel strategy against ischemia and reperfusion injury: cytoprotection with heme oxygenase system.

Masamichi Katori; Dean M. Anselmo; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

Much interest has recently been focused on the physiological/pathological role of the heme oxygenase (HO) system, the rate-limiting step in the conversion of heme, in inflammatory events. The HO system may be instrumental in mediating a number of cytoprotective effects, because of its end products, biliverdin, carbon monoxide (CO) and ferrous free iron (Fe2+). As each of the byproducts acts dependently and/or co-operatively with each other, their in vivo effects are complex. In general, the HO system is thought to exert three major functions in ischemia/reperfusion injury: (1) anti-oxidant effects; (2) maintenance of microcirculation; and (3) modulatory effects upon the cell cycle. The anti-oxidant functions depend on heme degradation, oxygen consumption and the production of biliverdin/ferritin via iron accumulation. On the other hand, the production of CO, which has vasodilatory and anti-platelet aggregative properties, can maintain tissue microcirculation. Strikingly, CO may also be instrumental in anti-apoptotic and cell arrest mechanisms. The HO system prevents early injury in the re-perfused organ, and inhibits the function of immune reactive cells, such as neutrophils, macrophages and lymphocytes. The role of the HO system as a novel strategy to mitigate an antigen-independent ischemia/reperfusion injury has been documented in a number of transplantation models.


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.


Liver Transplantation | 2007

A caspase inhibitor, IDN‐6556, ameliorates early hepatic injury in an ex vivo rat model of warm and cold ischemia

Niel C. Hoglen; Dean M. Anselmo; Masamichi Katori; Marian Kaldas; Xiu-Da Shen; Karen L. Valentino; Charles Lassman; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski; Douglas G. Farmer

This study examined the efficacy of the caspase inhibitor, IDN‐6556, in a rat model of liver ischemia‐reperfusion injury. Livers from male Sprague‐Dawley rats were reperfused for 120 minutes after 24 hours of 4°C cold storage in University of Wisconsin solution. Portal blood flow measurements estimated sinusoidal resistance, and bile production, alanine aminotransferase activities, and Suzuki scores were evaluated as parameters of hepatocyte/liver injury. Treated livers were exposed to 25 or 50 μM of IDN‐6556 in University of Wisconsin storage solution and/or the perfusate. All treatment regimens with IDN‐6556 significantly improved portal blood flow measured at 120 minutes, and significant improvements were seen as early as 30 minutes when inhibitor was also present in the perfusate (P < 0.01). All treatment groups with IDN‐6556 significantly increased bile production by 3‐4‐fold compared with controls (P < 0.01), and reductions in alanine aminotransferase activities were seen within 90 minutes of reperfusion (P < 0.05). These data were confirmed by improved Suzuki scores (less sinusoidal congestion, necrosis, and vacuolization) in all treated groups. Livers from the IDN‐6556–treated groups had markedly reduced caspase activities and TUNEL (terminal deoxynucleotidyl transferase dUTP nick‐end labeling)‐positive cells, suggesting reductions in apoptosis. IDN‐6556 present in cold storage media ameliorated liver injury due to cold ischemia and reperfusion injury and may be a rational therapeutic approach to reduce the risk of liver ischemia in the clinical setting. Liver Transpl 13: 361–366, 2007.


Transplantation | 2003

Interleukin-13 gene transfer protects rat livers from antigen-independent injury induced by ischemia and reperfusion.

Bibo Ke; Xiu-Da Shen; Charles Lassman; Feng Gao; Masamichi Katori; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

Background. Ischemia-reperfusion (I/R) injury is a prime inflammatory factor in the dysfunction of orthotopic liver transplants. Interleukin (IL)-13 suppresses macrophage production of proinflammatory mediators. This study explores the effects of adenovirus (Ad)-based IL-13 gene transfer in rat models of hepatic I/R injury. Methods. The authors used a model of warm in situ ischemia followed by reperfusion, and ex vivo cold ischemia followed by transplantation. Results. In a model of warm in situ ischemia followed by reperfusion, Ad-based IL-13 significantly diminished hepatocellular injury, assessed by serum glutamic oxaloacetic transaminase (SGOT) levels, as compared with Ad-based &bgr;-galactosidase (gal)-treated livers. In a model of ex vivo cold ischemia followed by transplantation, the survival of liver grafts increased from 50% in Ad-&bgr;-gal untreated controls to 100% after Ad-IL-13 gene therapy. This beneficial effect correlated with improved liver function (SGOT levels), preservation of hepatic histologic integrity and architecture (Suzuki criteria), and depression of neutrophil infiltration (myeloperoxidase assay). Ad-IL-13 diminished activation of macrophage-neutrophil–associated tumor necrosis factor-&agr;, macrophage inflammatory protein-2, and endothelial-dependent E-selectin, but increased type 2 IL-4 and IL-13 expression. Conclusions. This study documents striking cytoprotective effects of virally induced IL-13 against hepatic I/R injury in two clinically relevant rat models of hepatic I/R injury. These data provide the rationale for novel therapeutic approaches to maximize the organ donor pool through the safer use of liver transplants despite prolonged periods of warm or cold ischemia, or both.


Transplantation | 2000

Prior induction of heat shock proteins by a nitric oxide donor attenuates cardiac ischemia/reperfusion injury in the rat.

Masamichi Katori; Tohru Tamaki; Tsuyoshi Takahashi; Mitsuko Tanaka; Akio Kawamura; Akira Kakita

BACKGROUND Recent studies have demonstrated that nitric oxide (NO) releasers considerably increase heat shock proteins (HSPs) in the in vitro cell system, providing resistance to oxidant damage. This study was designed to examine the cellular responses of HSPs induced by prior administration of an NO releaser, FK409 (FK), in an in vivo transplantation model. METHODS Lewis rats received either saline or FK solution intravenously administered at different time points before graft harvesting (10 micromol/kg) or for 15 min during reperfusion (0.66 micromol/kg/min). Tissue specimens were taken to determine HSP70 and heme oxygenase-1/HSP32 (HO-1) expression, and glutathione content. After 24-hr preservation with University of Wisconsin solution, heterotopic cardiac transplantations were performed, and graft survival was determined at 14 days. Tissue samples for end labeling of nuclear DNA fragments (TdT-mediated d-uridine triphosphate biotin nick end labeling; TUNEL) and propidium iodide staining were taken 15 min after reperfusion. RESULTS The gene and protein expression of HSP70 after FK administration peaked at 12 min and 60-90 min, whereas those of HO-1 peaked at 6 min and 90 min, respectively. Then, representative cardiac grafts taken 60 min after FK treatment were examined for further assay. Localization of induced HSP70 and HO-1 molecules were observed in the myocardium and vascular endothelium, respectively. Prior treatment of FK was effective in preventing the reduction of tissue glutathione contents compared with control (P<0.05). Fewer TUNEL and propidium iodide-positive cells were also observed in the FK group (P<0.0005, vs. control). The graft survival rate was higher in the FK group (9/10 vs. 1/10 of control; P<0.001), whereas the groups either harvested 10 min after FK pretreatment or continuously infused for 15 min during reperfusion were inferior, similar to that of control. CONCLUSION Prior induction of HSP70 and HO-1 with a relatively low dose of FK administration attenuates ischemia and reperfusion injury, which was due to antioxidant and antiapoptotic activities augmented by such stress proteins. Thus, NO releasers as a pharmacological maneuver may provide an innovative approach for the prevention of ischemia and reperfusion injury.


Transplantation Proceedings | 2003

Biliverdin protects rat livers from ischemia/reperfusion injury.

Constantino Fondevila; Masamichi Katori; Charles Lassman; Ian C. Carmody; Ronald W. Busuttil; F.H Bach; Jerzy W. Kupiec-Weglinski

OBJECTIVE To explore putative cytoprotective functions of biliverdin during hepatic ischemia/reperfusion (I/R) injury in rat models. MATERIAL AND METHODS Male Sprague Dawley (SD) rat livers were harvested and stored for 24 hours at 4 degrees C in University of Wisconsin (UW) solution (n=18), and then perfused with blood for 2 hours on an isolated rat liver perfusion apparatus equipped for temperature (37 degrees C), pressure (13 cm H2O), and pH (7.3) maintenance. Biliverdin was added to the blood at concentrations of 10 and 50 micromol in two groups of six animals. Portal vein blood flow, bile production, and GOT/GPT levels were assessed serially. At the conclusion of the experiment, liver samples were collected for histologic evaluation using Suzuki criteria. RESULTS BV exerted protective effects against liver I/R injury. Adjunctive biliverdin improved portal venous blood flow (mL/min/g) from the beginning of reperfusion (1.33+/-0.17 versus 0.98+/-0.15; P<.001) and increased bile production (mL/g) as compared with the control group (3.40 versus 1.88; P<.003). I/R-induced hepatocellular damage as measured by GOT/GPT release (IU/L) was diminished in the biliverdin group (91 versus 171 and 46 versus 144, respectively; P<.0001). Improved liver function by biliverdin was accompanied by preservation of the histologic structure as assessed by Suzuki criteria (3.7+/-1.4 versus 6.8+/-0.8 in untreated controls; P<.005). CONCLUSIONS Biliverdin attenuates the ischemia/early reperfusion injury of rat liver grafts as assessed by hemodynamics, function, enzyme analysis, and histology. This study provides the rationale for novel therapeutic approaches using biliverdin to maximize the organ donor pool through the safer use of liver transplants despite prolonged periods of cold ischemia.


Journal of Immunology | 2001

Homing of In Vitro-Generated Donor Antigen-Reactive CD4+ T Lymphocytes to Renal Allografts Is α4β1 But Not αLβ2 Integrin Dependent

Markus H. Hammer; Yuan Zhai; Masamichi Katori; Thomas Ritter; Hans-Dieter Volk; Ana J. Coito; Jerzy W. Kupiec-Weglinski

The extravasation and sequestration of Ag-reactive T lymphocytes into vascularized organ allografts depend on a cascade of complex interactions among circulating lymphocytes, endothelial cells, and extracellular matrix proteins. Ag-activated donor-specific CD4 T cells are major initiators and effectors in the allograft rejection response. Interfering with the intragraft homing of activated CD4 T cells may represent a novel therapeutic approach in transplant recipients. We have developed a FACS-based short-term homing assay that allows tracing in vitro-generated Ag-reactive CD4 T cells after adoptive transfer in test rat recipients. Allospecific cell lines were preincubated with anti-α4β1 or anti-αLβ2 mAb, because of enhanced expression of both integrin receptors after alloactivation. The pretreated LewisBN lymphocytes were carboxyfluorescein diacetate succinimidyl ester labeled and adoptively transferred into Lewis rat recipients of Brown Norway kidney allografts. The injection of equal numbers of PKH-26-labeled untreated cells allowed quantitative comparison of both populations in the same animal. Ex vivo treatment with anti-α4β1 mAb diminished intragraft infiltration of adoptively transferred T cells by 85% in a donor-specific fashion. In contrast, treatment with anti-αLβ2 mAb did not affect intragraft cell sequestration. Hence, blocking α4β1 integrin interactions represents a novel strategy in preventing local intragraft recruitment of Ag-reactive CD4 T cells in transplant recipients.


Liver Transplantation | 2008

Tezosentan, a Novel Endothelin Receptor Antagonist, Markedly Reduces Rat Hepatic Ischemia and Reperfusion Injury in Three Different Models

Douglas G. Farmer; Fady M. Kaldas; Dean M. Anselmo; Masamichi Katori; Xiu-Da Shen; Charles Lassman; Marian Kaldas; Martine Clozel; Ronald W. Busuttil; Jerzy W. Kupiec-Weglinski

This study investigated the effects of dual endothelin (ET) receptor blockade in rat models of liver ischemia and reperfusion injury (IRI). Three models of IRI were used: (1) in vivo total hepatic warm ischemia with portal shunting for 60 minutes with control (saline) and treatment groups (15 mg/kg tezosentan intravenously prior to reperfusion), (2) ex vivo hepatic perfusion after 24 hours of cold storage in University of Wisconsin solution with control and treatment groups (10 mg/kg tezosentan in the perfusate), and (3) syngeneic liver transplantation (LT) after 24 hours of cold storage in University of Wisconsin solution with control and treatment groups (10 mg/kg tezosentan intravenously prior to reperfusion). Tezosentan treatment significantly improved serum transaminase and histology after IRI in all 3 models. This correlated with reduced vascular resistance, improved bile production, and an improved oxygen extraction ratio. Treatment led to a reduction in neutrophil infiltration and interleukin‐1 beta and macrophage inflammatory protein 2 production. A reduction in endothelial cell injury as measured by purine nucleoside phosphorylase was seen. Survival after LT was significantly increased with tezosentan treatment (90% versus 50%). In conclusion, this is the first investigation to examine dual receptor ET blockade in 3 models of hepatic IRI and the first to use the parenterally administered agent tezosentan. The results demonstrate that in both warm and cold IRI tezosentan administration improves sinusoidal hemodynamics and is associated with improved tissue oxygenation and reduced endothelial cell damage. In addition, reduced tissue inflammation, injury, and leukocyte chemotactic signaling were seen. These results provide compelling data for the further investigation of the use of tezosentan in hepatic IRI. Liver Transpl 14:1737–1744, 2008.

Collaboration


Dive into the Masamichi Katori's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bibo Ke

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana J. Coito

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiu-Da Shen

University of California

View shared research outputs
Top Co-Authors

Avatar

Dean M. Anselmo

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Feng Gao

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge