Network


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

Hotspot


Dive into the research topics where Yoko Mullen is active.

Publication


Featured researches published by Yoko Mullen.


PLOS ONE | 2009

Insulin gene expression is regulated by DNA methylation.

Akio Kuroda; Tibor A. Rauch; Ivan Todorov; Hsun Teresa Ku; Ismail H. Al-Abdullah; Fouad Kandeel; Yoko Mullen; Gerd P. Pfeifer; Kevin Ferreri

Background Insulin is a critical component of metabolic control, and as such, insulin gene expression has been the focus of extensive study. DNA sequences that regulate transcription of the insulin gene and the majority of regulatory factors have already been identified. However, only recently have other components of insulin gene expression been investigated, and in this study we examine the role of DNA methylation in the regulation of mouse and human insulin gene expression. Methodology/Principal Findings Genomic DNA samples from several tissues were bisulfite-treated and sequenced which revealed that cytosine-guanosine dinucleotide (CpG) sites in both the mouse Ins2 and human INS promoters are uniquely demethylated in insulin-producing pancreatic beta cells. Methylation of these CpG sites suppressed insulin promoter-driven reporter gene activity by almost 90% and specific methylation of the CpG site in the cAMP responsive element (CRE) in the promoter alone suppressed insulin promoter activity by 50%. Methylation did not directly inhibit factor binding to the CRE in vitro, but inhibited ATF2 and CREB binding in vivo and conversely increased the binding of methyl CpG binding protein 2 (MeCP2). Examination of the Ins2 gene in mouse embryonic stem cell cultures revealed that it is fully methylated and becomes demethylated as the cells differentiate into insulin-expressing cells in vitro. Conclusions/Significance Our findings suggest that insulin promoter CpG demethylation may play a crucial role in beta cell maturation and tissue-specific insulin gene expression.


American Journal of Transplantation | 2007

Mesenchymal Stem Cells Facilitate the Induction of Mixed Hematopoietic Chimerism and Islet Allograft Tolerance without GVHD in the Rat

S. Itakura; S. Asari; Jeffrey Rawson; T. Ito; Ivan Todorov; Chih-Pin Liu; N. Sasaki; Fouad Kandeel; Yoko Mullen

Induction of hematopoietic chimerism and subsequent donor‐specific immune tolerance via bone marrow transplantation is an ideal approach for islet transplantation to treat type‐1 diabetes. We examined the potential of mesenchymal stem cells (MSCs) in the induction of chimerism and islet allograft tolerance without the incidence of graft‐versus‐host disease (GVHD). Streptozotocin‐diabetic rats received a conditioning regimen consisting of antilymphocyte serum and 5 Gy total body irradiation, followed by an intraportal co‐infusion of allogeneic MSCs, bone marrow cells (BMCs) and islets. Although all the recipients rejected the islets initially, half of them developed stable mixed chimerism and donor‐specific immune tolerance, shown by the engraftment of donor skin and second‐set islet transplants and acute rejection of a third‐party skin. The engraftment of the primary islet allografts with stable chimerism was achieved by the addition of a 2‐week peritransplant administration of 15‐deoxyspergualin (DSG). Without MSCs, none of the recipients treated with DSG developed chimerism or reversal of diabetes. GVHD was not observed in any of the recipients infused with MSCs (0/15), whereas it occurred in 4/11 recipients without MSCs. These results indicate a potential use of MSCs for induction of hematopoietic chimerism and subsequent immune tolerance in clinical islet transplantation.


Biomaterials | 2013

Maintaining functional islets through encapsulation in an injectable saccharide-peptide hydrogel.

Sophia W. Liao; Jeffrey Rawson; Keiko Omori; Kohei Ishiyama; Davoud Mozhdehi; Alina R. Oancea; Taihei Ito; Zhibin Guan; Yoko Mullen

Islet transplantation offers a promising treatment for type 1 diabetes (T1D). However, a major hurdle in this treatment is the rapid loss of functional islets during culture and after transplantation. The liver site, currently utilized for transplantation, is suboptimal for achieving long-term insulin independence due to a rapid islet loss followed by a chronic decline in islet function after transplantation. Herein, we report a synthetic saccharide-peptide (SP) hydrogel that allows suspending islets in liquid and injecting for in situ polymerization without forming islet clumps, indicating its potential in extrahepatic islet transplantation. Inxa0vitro, rat islets in SP hydrogel maintained a 3D structure and high glucose-stimulated insulin release similar to that observed in freshly isolated islets for 4 weeks, while control islets cultured in suspension lost their 3D structure and insulin release responses by 2 weeks. Biocompatibility of SP hydrogel was shown by the absence of cytokine mRNA activation in peripheral blood mononuclear cells (PBMCs) exposed to hydrogel inxa0vitro and by the absence of cellular infiltrates in and around the hydrogel implanted subcutaneously. Syngeneic Lewis rat islets transplanted in SP hydrogel in various extrahepatic sites stained strongly for insulin, and more effectively reversed diabetes than unencapsulated islets when transplanted in an omental pocket. In conclusion, the SP hydrogel is non-cytotoxic and supports normal islet structure and function both inxa0vitro and inxa0vivo. Specifically, the ability of the hydrogel to separate individual islets after transplantation is important for maintaining their function inxa0vivo. This important property, combined with the versatility and biocompatibility, makes our SP hydrogel a promising synthetic scaffold that can facilitate transplantation of organized heterogeneous cells to preserve their micro-structure and function.


American Journal of Transplantation | 2007

Improvement of human islet cryopreservation by a p38 MAPK inhibitor.

Keiko Omori; Luis Valiente; Chris Orr; Jeffrey Rawson; Kevin Ferreri; Ivan Todorov; Ismail H. Al-Abdullah; S. Medicherla; A. A. Potter; G. F. Schreiner; Fouad Kandeel; Yoko Mullen

The activation of p38 mitogen‐activated protein kinase (MAPK) has been shown to cause ischemia/reperfusion injury of several organs used for transplantation and also to play a significant role in primary islet graft nonfunction. Activation of p38 MAPK may also occur during islet cryopreservation and thawing. In this study, a p38 MAPK inhibitor (p38IH) was applied to human islet cryopreservation to improve islet yield and quality after thawing. Under serum‐free conditions, human islets were cryopreserved, thawed and cultured using our standard procedures. Three types of solutions were tested: conventional RPMI1640 medium (RPMI), a newly developed islet cryopreservation solution (ICS), and ICS supplemented with a p38IH, SD‐282 (ICS‐p38IH). Activation or inhibition of p38 MAPK was demonstrated by the diminished phosphorylation of HSP27 substrate. Islet recovery on day 2 after thawing was highest with ICS‐p38IH and islet viability was not significantly different in the three groups. β Cell numbers and function were the highest in islets cryopreserved with ICS‐p38IH. Glucose‐stimulated human C‐peptide levels were 86% of that of the nonfrozen islets when measured 4 weeks after transplantation into NODscid mice. This improvement may provide an opportunity to establish islet banks and allow the use of cryopreserved islets for clinical transplantation.


Cell Transplantation | 2015

Human Pancreatic Islets Isolated From Donors With Elevated HbA1c Levels: Islet Yield and Graft Efficacy.

Meirigeng Qi; Brian McFadden; Luis Valiente; Keiko Omori; Shiela Bilbao; Jemily Juan; Jeffrey Rawson; Alina R. Oancea; Stephen Scott; Indu Nair; Kevin Ferreri; Yoko Mullen; Donald Dafoe; Mohamed Ei-Shahawy; Fouad Kandeel; Ismail H. Al-Abdullah

The aim of this study was to investigate the effects of elevated donor HbA1c levels (type 2 diabetes, T2D) on the islet yield and functionality postisolation. In this retrospective analysis, donors for islet isolations were classified into two groups: T2D group (HbA1c ≥ 6.5%, n = 18) and normal group (HbA1c < 6.5%, n = 308). Optimum pancreas digestion time (switch time) was significantly higher in the T2D group compared to the normal group (13.7 ± 1.2 vs. 11.7 ± 0.1 min, respectively, p = 0.005). Islet yields were significantly lower in the T2D group compared to the control (T2D vs. control): islet equivalent (IEQ)/g (prepurification 2,318 ± 195 vs. 3,713 ± 114, p = 0.003; postpurification 1,735 ± 175 vs. 2,663 ± 89, p = 0.013) and islet particle number (IPN)/g (prepurification, 2,519 ± 336 vs. 4,433 ± 143, p = 0.001; postpurification, 1,760 ± 229 vs. 2,715 ± 85, p = 0.007). Islets from T2D pancreata had significantly lower viability (T2D vs. control: 91.9 ± 1.6 vs. 94.4 ± 0.3%, p = 0.004) and decreased oxygen consumption rate (DOCR) (T2D vs. control: 0.09 ± 0.01 and 0.21 ± 0.03 nmol O2 100 islets−1 min−1, p = 0.049). The islets isolated from T2D donor pancreata reversed diabetes in NOD-SCID mice in 9% (2/22) compared to islets from control donor pancreata, which reversed diabetes in 67% (175/260, p < 0.001). In conclusion, this study demonstrates that elevated HbA1c (≥6.5%) is associated with impairment of islet function and lower islet yield; however, these islets could not be suitable for clinical applications.


Immunology | 2005

Identification of the VH genes encoding xenoantibodies in non‐immunosuppressed rhesus monkeys

Annette Kleihauer; Clare R. Gregory; Dominic C. Borie; Andrew E. Kyles; Irina Shulkin; Insiyyah Patanwala; Joanne Zahorsky-Reeves; Vaughn A. Starnes; Yoko Mullen; Ivan Todorov; Mary Kearns-Jonker

The major immunological barrier that prevents the use of wild‐type pig xenografts as an alternative source of organs for human xenotransplantation is antibody‐mediated rejection. In this study, we identify the immunoglobulin variable region heavy (IgVH) chain genes encoding xenoantibodies to porcine heart and fetal porcine islet xenografts in non‐immunosuppressed rhesus monkeys. We sought to compare the IgVH genes encoding xenoantibodies to porcine islets and solid organ xenografts. The immunoglobulin M (IgM) and IgG xenoantibody response was analysed by enzyme‐linked immunosorbent assay and cDNA libraries from peripheral blood lymphocytes were prepared and sequenced. The relative frequency of IgVH gene usage was established by colony filter hybridization. Induced xenoantibodies were encoded by the IGHV3‐11 germline progenitor, the same germline gene that encodes xenoantibodies in humans mounting active xenoantibody responses. The immune response to pig xenografts presented as solid organs or isolated cells is mediated by identical IgVH genes in rhesus monkeys. These animals represent a clinically relevant model to identify the immunological basis of pig‐to‐human xenograft rejection.


Biochemical and Biophysical Research Communications | 2016

Isolated human islets require hyperoxia to maintain islet mass, metabolism, and function.

Hirotake Komatsu; Dongyang Kang; Leonard Medrano; Alyssa Barriga; Daniel Mendez; Jeffrey Rawson; Keiko Omori; Kevin Ferreri; Yu-Chong Tai; Fouad Kandeel; Yoko Mullen

Pancreatic islet transplantation has been recognized as an effective treatment for Type 1 diabetes; however, there is still plenty of room to improve transplantation efficiency. Because islets are metabolically active they require high oxygen to survive; thus hypoxia after transplant is one of the major causes of graft failure. Knowing the optimal oxygen tension for isolated islets would allow a transplant team to provide the best oxygen environment during pre- and post-transplant periods. To address this issue and begin to establish empirically determined guidelines for islet maintenance, we exposed inxa0vitro cultured islets to different partial oxygen pressures (pO2) and assessed changes in islet volume, viability, metabolism, and function. Human islets were cultured for 7 days in different pO2 media corresponding to hypoxia (90xa0mmHg), normoxia (160xa0mmHg), and hyerpoxia (270 or 350xa0mmHg). Compared to normoxia and hypoxia, hyperoxia alleviated the loss of islet volume, maintaining higher islet viability and metabolism as measured by oxygen consumption and glucose-stimulated insulin secretion responses. We predict that maintaining pre- and post-transplanted islets in a hyperoxic environment will alleviate islet volume loss and maintain islet quality thereby improving transplant outcomes.


Diabetologia | 2011

mRNA of the pro-apoptotic gene BBC3 serves as a molecular marker for TNF-α-induced islet damage in humans

Keiko Omori; M. Mitsuhashi; K. Ishiyama; Indu Nair; Jeffrey Rawson; Ivan Todorov; Fouad Kandeel; Yoko Mullen

Aims/hypothesisTNF-α plays important roles in the pathogenesis of type 1 and type 2 diabetes mellitus. In light of this, we examined the involvement of a pro-apoptotic gene, BBC3 (also known as PUMA), in TNF-α-mediated beta cell dysfunction and destruction in human islets.MethodsHuman islets were exposed in vitro to TNF-α alone or in combination with IFN-γ. Gene expression was assessed by RT-PCR using a set of single islets. Protein abundance and cellular localisation of BBC3 were assessed by immunoblot and immunohistochemistry. A marginal number of islets were transplanted into diabetic NODscid mice to correlate in vivo islet function with BBC3 expression.ResultsBBC3 and IL8 mRNA were upregulated in TNF-α-stimulated islets in a dose-dependent manner and enhanced through addition of IFN-γ, but not upregulated by IFN-γ alone. Immunohistochemistry revealed that TNF-α in combination with IFN-γ upregulated basal BBC3 abundance in the cytoplasm of beta cells along with the perinuclear clustering of mitochondria partially co-localised with BBC3. TNF-α alone did not induce beta cell death, but did abrogate preproinsulin precursor mRNA synthesis in response to high glucose stimulation, which was inversely associated with upregulation of BBC3 mRNA expression by TNF-α. Higher BBC3 mRNA expression in islets correlated with decreased graft function in vivo.Conclusions/interpretationThese results suggest that BBC3 mRNA can serve as a molecular marker to detect early TNF-α-induced beta cell stress and may help identify islet-protective compounds for the treatment of diabetes.


American Journal of Physiology-endocrinology and Metabolism | 2015

Sodium levels of human pancreatic donors are a critical factor for determination of islet efficacy and survival

Meirigeng Qi; Valiente Luis; Shiela Bilbao; Keiko Omori; Jeffrey Rawson; Brian McFadden; Jemily Juan; Indu Nair; Yoko Mullen; Mohamed El-Shahawy; Donald Dafoe; Fouad Kandeel; Ismail H. Al-Abdullah

Organs from hypernatremia (elevated Na+) donors when used for transplantation have had dismal outcomes. However, islet isolation from hypernatremic donors for both transplantation and research applications has not yet been investigated. A retrospective analysis of in vivo and in vitro islet function studies was performed on islets isolated from hypernatremic (serum sodium levels≥160 meq/l) and normal control (serum sodium levels≤155 meq/l) donors. Twelve isolations from 32 hypernatremic and 53 isolations from 222 normal donors were randomly transplanted into diabetic NOD Scid mice. Sodium levels upon pancreas procurement were significantly elevated in the hypernatremia group (163.5±0.6 meq/l) compared with the normal control group (145.9±0.4 meq/l) (P<0.001). The postculture islet recovery rate was significantly lower in the hypernatremia (59.1±3.8%) group compared with the normal (73.6±1.8%) group (P=0.005). The duration of hypernatremia was inversely correlated with the recovery rate (r2=0.370, P<0.001). Furthermore, the percentage of successful graft function when transplanted into diabetic NOD Scid mice was significantly lower in the hypernatremia (42%) group compared with the normal control (85%) group (P<0.001). The ability to predict islet graft function posttransplantation using donor sodium levels and duration of hypernatremia was significant (ROC analysis, P=0.022 and 0.042, respectively). In conclusion, duration of donor hypernatremia is associated with reduced islet recovery postculture. The efficacy of islets from hypernatremia donors diminished when transplanted into diabetic recipients.


Cryobiology | 2016

Mechanisms of islet damage mediated by pancreas cold ischemia/rewarming

Keiko Omori; Eiji Kobayashi; Jeffrey Rawson; Masafumi Takahashi; Yoko Mullen

Prolonged pancreas cold ischemia is known to negatively correlate with islet isolation outcomes, hindering successful islet transplantation to treat Type-1 Diabetes. Due to poor islet isolation outcome, pancreata with over 16xa0h cold ischemia are currently not considered for islet transplantation. Mechanisms involved in pancreas cold ischemia/rewarming mediated islet damage during islet isolation and culture are not well understood. Using an en bloc cold preserved rat pancreas preparation, we attempted to clarify possible mechanisms of islet death associated with prolonged pancreas cold ischemia and subsequent rewarming. Cold ischemia lasting 16xa0h decreased post-isolation islet yield and increased islet death during the initial 6xa0h of culture. Electron micrographs revealed swelling and severe disruption of cellular and mitochondrial membranes, as well as an enlarged endoplasmic reticulum (ER) in β-cells isolated from cold preserved pancreata. Prolonged cold ischemia of the pancreas transiently activated mitogen-activated protein kinases (MAPKs) in isolated islets and increased lipid peroxidation products 4-hydroxynonenal (HNE) and heat shock protein (Hsp) 70 after culture, indicating the activation of oxidative stress signaling pathways. The islet isolation process, irrespective of pancreas cold ischemia, activated unfolded protein response (UPR), while the ER protective chaperon BiP was further upregulated by pancreas cold ischemia/rewarming. During the first 6xa0h of culture following islet isolation, p53 upregulated modulator of apoptosis (Puma) and caspase-3 activation were also upregulated. Our study indicates the involvement of both apoptosis and necrosis in islet death, and suggests oxidative stress and disruption of membranes are critical mechanisms mediated by pancreas cold ischemia/rewarming.

Collaboration


Dive into the Yoko Mullen's collaboration.

Top Co-Authors

Avatar

Fouad Kandeel

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey Rawson

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Keiko Omori

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ismail H. Al-Abdullah

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ivan Todorov

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kevin Ferreri

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hirotake Komatsu

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Luis Valiente

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chris Orr

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Indu Nair

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge