Network


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

Hotspot


Dive into the research topics where Gretchen A. LaRusch is active.

Publication


Featured researches published by Gretchen A. LaRusch.


Cancer Research | 2007

Nutlin3 Blocks Vascular Endothelial Growth Factor Induction by Preventing the Interaction between Hypoxia Inducible Factor 1α and Hdm2

Gretchen A. LaRusch; Mark W. Jackson; James D. Dunbar; Robert S. Warren; David B. Donner; Lindsey D. Mayo

Hdm2 is elevated in numerous types of malignancies and is thought to impede the function of wild-type p53. Reactivation of p53 by disrupting the association with Hdm2 was the impetus for the development of Nutlin3. Although regulation of p53 has been the central focus of Hdm2 activity, it also binds other proteins through its p53-binding domain. Here, we show that hypoxia-inducible factor 1alpha (HIF1alpha) binds to Hdm2 in the domain designated to bind p53. HIF1alpha and p53 share a conserved motif that is required to bind Hdm2. Distinct complexes form between Hdm2-HIF1alpha and Hdm2-p53 as determined by immunoprecipitation of nuclear extracts and in vitro. The Hdm2 antagonist Nutlin3 prevents the association between Hdm2 and HIF1alpha. The vascular endothelial growth factor (VEGF) gene is a transcriptional target of HIF1alpha, and under normoxic or hypoxic conditions, Hdm2 increases HIF1alpha activity to induce VEGF production. Blocking the association of Hdm2 and HIF1alpha by Nutlin3, or ablating Hdm2 expression, diminished the level of VEGF under conditions of normoxia or hypoxia. Our findings establish a unique role for Nutlin3 in attenuating VEGF induction by preventing the association of Hdm2 with HIF1alpha.


Science | 2015

Inhibition of the prostaglandin-degrading enzyme 15-PGDH potentiates tissue regeneration

Yongyou Zhang; Amar Desai; Sung Yeun Yang; Ki Beom Bae; Monika I. Antczak; Stephen P. Fink; Shruti Tiwari; Joseph Willis; Noelle S. Williams; Dawn M. Dawson; David Wald; Wei Dong Chen; Zhenghe Wang; Lakshmi Kasturi; Gretchen A. LaRusch; Lucy He; Fabio Cominelli; Luca Di Martino; Zora Djuric; Ginger L. Milne; Mark R. Chance; Juan R. Sanabria; Chris Dealwis; Debra Mikkola; Jacinth Naidoo; Shuguang Wei; Hsin Hsiung Tai; Stanton L. Gerson; Joseph M. Ready; Bruce A. Posner

A shot in the arm for damaged tissue Tissue damage can be caused by injury, disease, and even certain medical treatments. There is great interest in identifying drugs that accelerate tissue regeneration and recovery, especially drugs that might benefit multiple organ systems. Zhang et al. describe a compound with this desired activity, at least in mice (see the Perspective by FitzGerald). SW033291 promotes recovery of the hematopoietic system after bone marrow transplantation, prevents the development of ulcerative colitis in the intestine, and accelerates liver regeneration after hepatic surgery. It acts by inhibiting an enzyme that degrades prostaglandins, lipid signaling molecules that have been implicated in tissue stem cell maintenance. Science, this issue 10.1126/science.aaa2340; see also p. 1208 A compound that inhibits prostaglandin degradation enhances tissue regeneration in multiple organs in mice. [Also see Perspective by FitzGerald] INTRODUCTION Agents that promote tissue regeneration could be beneficial in a variety of clinical settings, such as stimulating recovery of the hematopoietic system after bone marrow transplantation. Prostaglandin PGE2, a lipid signaling molecule that supports expansion of several types of tissue stem cells, is a candidate therapeutic target for promoting tissue regeneration in vivo. To date, therapeutic interventions have largely focused on targeting two PGE2 biosynthetic enzymes, cyclooxygenase-1 and cyclooxygenase-2 (COX-1 and COX-2), with the aim of reducing PGE2 production. In this study, we take the converse approach: We examine the role of a prostaglandin-degrading enzyme, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), as a negative regulator of tissue repair, and we explore whether inhibition of this enzyme can potentiate tissue regeneration in mouse models. RATIONALE We used 15-PGDH knockout mice to elucidate the role of 15-PGDH in regulating tissue levels of PGE2 and tissue repair capacity in multiple organs. We then developed SW033291, a potent small-molecule inhibitor of 15-PGDH with activity in vivo. We used SW033291 to investigate the therapeutic potential of 15-PGDH inhibitors in tissue regeneration and to identify a 15-PGDH–regulated hematopoietic pathway within the bone marrow niche. RESULTS We found that in comparison with wild-type mice, 15-PGDH–deficient mice display a twofold increase in PGE2 levels across multiple tissues—including bone marrow, colon, and liver—and that they show increased fitness of these tissues in response to damage. The mutant mice also show enhanced hematopoietic capacity, with increased neutrophils, increased bone marrow SKL (Sca-1+ C-kit+ Lin−) cells (enriched for stem cells), and greater capacity to generate erythroid and myeloid colonies in cell culture. The 15-PGDH–deficient mice respond to colon injury from dextran sulfate sodium (DSS) with a twofold increase in cell proliferation in colon crypts, which confers resistance to DSS-induced colitis. The mutant mice also respond to partial hepatectomy with a greater than twofold increase in hepatocyte proliferation, which leads to accelerated and more extensive liver regeneration. SW033291, a potent small-molecule inhibitor of 15-PGDH (inhibitor dissociation constant Ki ~0.1 nM), recapitulates in mice the phenotypes of 15-PGDH gene knockout, inducing increased hematopoiesis, resistance to DSS colitis, and more rapid liver regeneration after partial hepatectomy. Moreover, SW033291-treated mice show a 6-day-faster reconstitution of hematopoiesis after bone marrow transplantation, with accelerated recovery of neutrophils, platelets, and erythrocytes, and greater recovery of bone marrow SKL cells. This effect is mediated by bone marrow CD45– cells, which respond to increased PGE2 with a fourfold increase in production of CXCL12 and SCF, two cytokines that play key roles in hematopoietic stem cell homing and maintenance. CONCLUSIONS Studying mouse models, we have shown that 15-PGDH negatively regulates tissue regeneration and repair in the bone marrow, colon, and liver. Of most direct utility, our observations identify 15-PGDH as a therapeutic target and provide a chemical formulation, SW033291, that is an active 15-PGDH inhibitor in vivo and that potentiates repair in multiple tissues. SW033291 or related compounds may merit clinical investigation as a strategy to accelerate recovery after bone marrow transplantation and other tissue injuries. Inhibiting 15-PGDH accelerates tissue repair. (A) The enzyme 15-PGDH degrades and negatively regulates PGE2. (B) SW033291 inhibits 15-PGDH, increases tissue levels of PGE2, and induces CXCL12 and SCF expression from CD45– bone marrow cells. This in turn accelerates homing of transplanted hematopoietic stem cells (HSC), generation of mature blood elements, and post-transplant recovery of normal blood counts. Inhibiting 15-PGDH similarly stimulates cell proliferation after injury to colon or liver, accelerating repair of these tissues. Agents that promote tissue regeneration could be beneficial in a variety of clinical settings, such as stimulating recovery of the hematopoietic system after bone marrow transplantation. Prostaglandin PGE2, a lipid signaling molecule that supports expansion of several types of tissue stem cells, is a candidate therapeutic target for promoting tissue regeneration in vivo. Here, we show that inhibition of 15-hydroxyprostaglandin dehydrogenase (15-PGDH), a prostaglandin-degrading enzyme, potentiates tissue regeneration in multiple organs in mice. In a chemical screen, we identify a small-molecule inhibitor of 15-PGDH (SW033291) that increases prostaglandin PGE2 levels in bone marrow and other tissues. SW033291 accelerates hematopoietic recovery in mice receiving a bone marrow transplant. The same compound also promotes tissue regeneration in mouse models of colon and liver injury. Tissues from 15-PGDH knockout mice demonstrate similar increased regenerative capacity. Thus, 15-PGDH inhibition may be a valuable therapeutic strategy for tissue regeneration in diverse clinical contexts.


Blood | 2011

Murine prolylcarboxypeptidase depletion induces vascular dysfunction with hypertension and faster arterial thrombosis

Gregory N. Adams; Gretchen A. LaRusch; Evi X. Stavrou; Yihua Zhou; Marvin T. Nieman; Gretta H. Jacobs; Yingjie Cui; Yuan Lu; Mukesh K. Jain; Fakhri Mahdi; Zia Shariat-Madar; Yoshio Okada; Louis G. D'Alecy; Alvin H. Schmaier

Prolylcarboxypeptidase (PRCP) activates prekallikrein to plasma kallikrein, leading to bradykinin liberation, and degrades angiotensin II. We now identify PRCP as a regulator of blood vessel homeostasis. β-Galactosidase staining in PRCP(gt/gt) mice reveals expression in kidney and vasculature. Invasive telemetric monitorings show that PRCP(gt/gt) mice have significantly elevated blood pressure. PRCP(gt/gt) mice demonstrate shorter carotid artery occlusion times in 2 models, and their plasmas have increased thrombin generation times. Pharmacologic inhibition of PRCP with Z-Pro-Prolinal or plasma kallikrein with soybean trypsin inhibitor, Pro-Phe-Arg-chloromethylketone or PKSI 527 also shortens carotid artery occlusion times. Aortic and renal tissues have uncoupled eNOS and increased reactive oxygen species (ROS) in PRCP(gt/gt) mice as detected by dihydroethidium or Amplex Red fluorescence or lucigenin luminescence. The importance of ROS is evidenced by the fact that treatment of PRCP(gt/gt) mice with antioxidants (mitoTEMPO, apocynin, Tempol) abrogates the hypertensive, prothrombotic phenotype. Mechanistically, our studies reveal that PRCP(gt/gt) aortas express reduced levels of Kruppel-like factors 2 and 4, thrombomodulin, and eNOS mRNA, suggesting endothelial cell dysfunction. Further, PRCP siRNA treatment of endothelial cells shows increased ROS and uncoupled eNOS and decreased protein C activation because of thrombomodulin inactivation. Collectively, our studies identify PRCP as a novel regulator of vascular ROS and homeostasis.


Journal of Biological Chemistry | 2006

Hdm2 Nuclear Export, Regulated by Insulin-like Growth Factor-I/MAPK/p90Rsk Signaling, Mediates the Transformation of Human Cells

Mark W. Jackson; Linnea E. Patt; Gretchen A. LaRusch; David B. Donner; George R. Stark; Lindsey D. Mayo

Insulin-like growth factor (IGF)-I receptor activation leads to enhanced proliferation and cell survival via the MAP kinase and phosphatidylinositol 3-kinase-signaling pathways. Upon stimulation by IGF-I, the Hdm2 oncoprotein is phosphorylated by AKT, leading to its rapid nuclear translocation and subsequent inhibition of p53. We now show that IGF-I stimulation regulates the nuclear export of Hdm2 and p53 via the MAP kinase pathway. Inhibition of p38 MAPK or MEK via pharmacological means or expression of dominant negative proteins inhibited the cytoplasmic accumulation of Hdm2 and increased Hdm2 and p53 protein levels, whereas constitutively active p90Rsk promoted the nuclear export of Hdm2. Expression of constitutively active p90Rsk with E1A, oncogenic H-Ras, and hTERT resulted in the anchorage-independent growth of normal human fibroblasts. Our findings link p90Rsk-mediated modulation of Hdm2 nuclear to cytoplasmic shuttling with the diminished ability of p53 to regulate cell cycle checkpoints that ultimately leads to transformation.


Blood | 2013

Prolylcarboxypeptidase promotes angiogenesis and vascular repair

Gregory N. Adams; Evi X. Stavrou; Chao Fang; Alona Merkulova; M. Amer Alaiti; Kohsuke Nakajima; Toshifumi Morooka; Sergei Merkulov; Gretchen A. LaRusch; Daniel I. Simon; Mukesh K. Jain; Alvin H. Schmaier

Prolylcarboxypeptidase (PRCP) is associated with leanness, hypertension, and thrombosis. PRCP-depleted mice have injured vessels with reduced Kruppel-like factor (KLF)2, KLF4, endothelial nitric oxide synthase (eNOS), and thrombomodulin. Does PRCP influence vessel growth, angiogenesis, and injury repair? PRCP depletion reduced endothelial cell growth, whereas transfection of hPRCP cDNA enhanced cell proliferation. Transfection of hPRCP cDNA, or an active site mutant (hPRCPmut) rescued reduced cell growth after PRCP siRNA knockdown. PRCP-depleted cells migrated less on scratch assay and murine PRCP(gt/gt) aortic segments had reduced sprouting. Matrigel plugs in PRCP(gt/gt) mice had reduced hemoglobin content and angiogenic capillaries by platelet endothelial cell adhesion molecule (PECAM) and NG2 immunohistochemistry. Skin wounds on PRCP(gt/gt) mice had delayed closure and reepithelialization with reduced PECAM staining, but increased macrophage infiltration. After limb ischemia, PRCP(gt/gt) mice also had reduced reperfusion of the femoral artery and angiogenesis. On femoral artery wire injury, PRCP(gt/gt) mice had increased neointimal formation, CD45 staining, and Ki-67 expression. Alternatively, combined PRCP(gt/gt) and MRP-14(-/-) mice were protected from wire injury with less neointimal thickening, leukocyte infiltration, and cellular proliferation. PRCP regulates cell growth, angiogenesis, and the response to vascular injury. Combined with its known roles in blood pressure and thrombosis control, PRCP is positioned as a key regulator of vascular homeostasis.


Thrombosis and Haemostasis | 2010

Oral thrombostatin FM19 inhibits prostate cancer.

Marvin T. Nieman; Gretchen A. LaRusch; Chao Fang; Yihua Zhou; Alvin H. Schmaier

Thrombin stimulates proliferation, invasion and metastasis by cleaving protease-activated receptor 1 (PAR1) on human prostate cancer cells. Current direct thrombin inhibitors pose risks for bleeding in the cancer patients. We have developed an oral reversible direct thrombin inhibitor called FM19. FM19 inhibits thrombin-induced calcium mobilisation of PC3 cells with an IC50 of 15 μM with a 95% confidence interval of 7.3-31.6 μM. Thrombin stimulation increases PC3 cell invasion three-fold from 27.1 ± 11.4 to 66 ± 11.6. FM19 or bivalirudin reduces cell invasion at ≥0.1 μM (p≤0.02). After inoculation with PC3 cells, nude mice were treated with oral FM19 at 3 mg/ml in the drinking water. The treated mice did not have long bleeding times and only a 1.4-fold increase in their thrombin clotting time. However, with treatment, the mice have a reduced rate of tumour growth 0.26 ± 0.17 fold change/day vs. 0.55 ± 0.35 for untreated (p = 0.038), reduced fold change in tumour size 5.3 ± 0.47 to 8.9 ± 1.8 (untreated) (p=0.048), and reduced overall tumour weight 0.5 ± 0.31 g vs. 0.82 ± 0.32 g (untreated) (p=0.04). On microscopic examination, FM19 treatment reduces the number of large vessels in the tumours from 4.6 ± 2.1 per high-powered field in untreated samples to 1.4 ± 1.4 in treated samples (p≤0.04). These studies show FM19 reduces prostate tumour growth in vivo at a concentration below that needed for anticoagulation. These data suggest novel opportunities for oral direct thrombin inhibitors in cancer therapy.


Thrombosis and Haemostasis | 2010

Factor XII: New life for an old protein

Alvin H. Schmaier; Gretchen A. LaRusch

Ratnoff and his coworkers recognised that factor XII (XII) stimulates cell growth and activates mitogen-activated protein kinase. We determined the receptor(s) for this function and the consequence of this signalling pathway. Investigations show that the urokinase plasminogen activator receptor serves as the XII binding site on cultured umbilical vein endothelial cells. When XII binds, it stimulates ERK1/2 and Akt S473 phosphorylation. These events are distinct because when cell mTORC2 is absent, XII phosphorylates ERK1/2 but not Akt S473. Zymogen XII is an equal stimulator of signalling as XIIa or inhibitor-treated XIIa. Peptides from uPAR domain 2 block XII binding and ERK1/2 and Akt phosphorylation. Furthermore, antibodies to the integrins β1 and α5 block XII signalling. Likewise, inhibitors to the EGFR block XII-induced phosphorylation events. XII stimulates cell growth and proliferation. XII induces angiogenesis ex vivo in normal aortic sprouts and in vivo in matrigel plugs in normal mice, but not in aorta from uPAR knockout mice or matrigel plugs placed into uPAR-deleted mice. Skin biopsies constitutively or in a wound nine days after injury show reduced CD31 antigen expression in specimens from XII knockout mice compared to wild-type mice. These studies indicate that XII stimulates angiogenesis, a physiologic function independent of contact activation.


American Journal of Physiology-heart and Circulatory Physiology | 2013

Domain 2 of uPAR regulates single-chain urokinase-mediated angiogenesis through β1-integrin and VEGFR2

Gretchen A. LaRusch; Alona Merkulova; Fakhri Mahdi; Zia Shariat-Madar; Robert G. Sitrin; Douglas B. Cines; Alvin H. Schmaier

How single-chain urokinase (ScuPA) mediates angiogenesis is incompletely understood. ScuPA (≥4 nM) induces phosphorylated (p)ERK1/2 (MAPK44 and MAPK42) and pAkt (Ser(473)) in umbilical vein and dermal microvascular endothelial cells. Activation of pERK1/2 by ScuPA is blocked by PD-98059 or U-0126, and pAkt (Ser(473)) activation is inhibited by wortmannin or LY-294002. ScuPA (32 nM) or protease-inhibited two-chain urokinase stimulates pERK1/2 to the same extent, indicating that signaling is not dependent on enzymatic activity. ScuPA induces pERK1/2, but not pAkt (Ser(473)), in SIN1(-/-) cells, indicating that the two pathways are not identical. Peptides from domain 2 of the urokinase plasminogen activator receptor (uPAR) or domain 5 of high-molecular-weight kininogen compete with ScuPA for the induction of pERK1/2 and pAkt (Ser(473)). A peptide of the integrin-binding site on uPAR, a β1-integrin peptide that binds uPAR, antibody 6S6 to β1-integrin, tyrosine kinase inhibitors AG-1478 or PP3, and small interfering RNA knockdown of VEFG receptor 2, but not HER1-HER4, blocked ScuPA-induced pERK1/2 and pAkt (Ser(473)). ScuPA-induced endothelial cell proliferation was blocked by inhibitors of pERK1/2 and pAkt (Ser(473)), antibody 6S6, and uPAR or kininogen peptides. ScuPA initiated aortic sprouts and Matrigel plug angiogenesis in normal, but not uPAR-deficient, mouse aortae or mice, respectively, but these were blocked by PD-98059, LY-294002, AG-1478, or cleaved high-molecular-weight kininogen. In summary, this investigation indicates a novel, a nonproteolytic signaling pathway initiated by zymogen ScuPA and mediated by domain 2 of uPAR, β1-integrins, and VEGF receptor 2 leading to angiogenesis. Kininogens or peptides from it downregulate this pathway.


Journal of Thrombosis and Haemostasis | 2011

Factor XII gene mutation in the Hageman family.

Y. O. Xu-Cai; J. Shen; S. Chen; Y. Zhou; Gretchen A. LaRusch; Evi X. Stavrou; Alvin H. Schmaier; Qingyu Wu

Hageman trait refers to an inherited deficiency of coagulation factor XII (FXII). The term was named by Oscar Ratnoff in 1955 after the index patient John Hageman, who had ‘incoagulable’ blood in vitro but no abnormal bleeding even after surgeries [1, 2]. Subsequently, Earl Davie and Oscar Ratnoff isolated Hageman factor, the protein missing in Hageman’s blood, which later was called FXII [3, 4]. This pioneer work contributed to the ‘Waterfall/Cascade’ hypothesis of blood coagulation presented in 1964 [5, 6], which established the basic principle that blood coagulation is mediated by the sequential activation of a series of plasma proteases. John Hageman died of pulmonary thromboembolism during bed rest from a broken hip in 1968 before gene cloning techniques were available [7]. As a result, the genetic defect underlying his FXII-deficiency was never determined. In this study, we obtained blood samples from descendants of the Hageman family to analyze their F12 gene that encodes FXII. The study was approved by the Cleveland Clinic Institutional Review Board. All participants provided written consent. John Hageman was never married and had no known descendants (Fig. 1A). We obtained blood samples from III-1, III-3-5 and IV-3-5, who were family members of his siblings’ descendants. None of these individuals had a history of abnormal bleeding. PCR and DNA sequencing of all 14 F12 gene exons in these individuals found no deletion, insertion or non-synonymous point mutations. In III-3 and IV-5, however, a G→A intronic mutation was identified at nucleotide position 11396 of the F12 gene [8] (Fig. 1B), which was part of the splice acceptor site of exon 14 (Fig. 1C). The mutation abolished the original splice acceptor site and created a new site one nucleotide downstream. As a result, the exon 14 reading frame was shifted by one position, thereby encoding a new peptide that replaced the catalytic serine and the rest of the protease domain (Fig. 1C). Fig. 1 (A) Hageman family pedigree. The pedigree was modified for confidentiality. J.H., John Hageman. Diamonds with a slash line represent deceased individuals. Diamonds with a dot represent heterozygous carriers. IV-1–2 were not available for the study. ... Interestingly, this 11396(G→A) mutation was reported previously in FXII-deficient individuals in Germany [9, 10]. PCR analysis of transcripts from the mutant F12 gene confirmed the predicted fusion mRNA. The study also showed that the fusion protein, if translated from the fusion mRNA, was unstable, because individuals homozygous for this mutation had no detectable plasma FXII antigen or activity [9, 10]. By ELISA (Innovative Research Inc.), we found that plasma FXII antigen levels in III-3 and IV-5, who were heterozygous for the mutation, were ~50% of that in other members of the family (Fig. 1A). The antigen level in the normal individuals of this family was ~1.8 fold higher than that of pooled normal plasma. The levels in III-3 and IV-5 were at ~90% of pooled normal plasma level (data not shown). As measured by a modified one-stage activated partial thromboplastin time, plasma FXII coagulant activity in four family members (III-1 and IV-3-5), whose blood was collected in sodium citrate, were 1.31, 1.44, 1.28 and 1.21 U/mL, respectively. On immunoblots, plasma FXII protein in all samples had a similar molecular mass, although the level in samples from III-3 and IV-5 was lower (data not shown). No additional FXII protein bands were detected on immunoblots. The data were consistent with the genotype in these individuals. These studies identified an F12 gene mutation in the descendants of the Hageman family. We showed that there was a mutant allele in III-3 and IV-5, which was likely from II-3 who inherited it from I-1 or I-2. It is likely that John Hageman (II-1) also inherited this mutant allele. Given the fact that there was no detectable FXII activity in his blood [2], John Hageman was expected to be either homozygous for this mutant allele or compounded heterozygous with another unknown mutant F12 allele. An inquiry of family history indicated no evidence that his parents were consanguineous but revealed that they emigrated from the central part of Germany, where the same mutant F12 allele was reported in at least five unrelated families [9, 10]. This mutant allele also existed in another unrelated family in Switzerland [10]. All of the individuals with this allele were asymptomatic and identified only by chance during hospital visits. Apparently, this mutation originated for sometime in history and is present in a number of individuals in Europe, especially in Germany. Since no other F12 mutations were found in the descendents of his family, the possibility cannot be excluded that John Hageman may have inherited two copies of the same mutant F12 allele from his parents. The chance encounter of John Hageman by Oscar Ratnoff more than half a century ago had a major role in the history of understanding the blood coagulation system [1, 3, 11–14]. Now FXII is known to be important in the contact activation system to regulate blood pressure, vascular permeability, complement activation, inflammatory responses, angiogenesis, and thrombosis risk [15–17]. Our finding of the F12 gene mutation in the Hageman family, providing an explanation to the long-standing question of John Hageman’s genetic defects, adds a fresh footnote to this important history in hematology.


Haematologica | 2018

A second-generation 15-PGDH inhibitor promotes bone marrow transplant recovery independently of age, transplant dose and granulocyte colony-stimulating factor support

Amar Desai; Yongyou Zhang; Youngsoo Park; Dawn M. Dawson; Gretchen A. LaRusch; Lakshmi Kasturi; David Wald; Joseph M. Ready; Stanton L. Gerson; Sanford D. Markowitz

Hematopoietic stem cell transplantation following myeloablative chemotherapy is a curative treatment for many hematopoietic malignancies. However, profound granulocytopenia during the interval between transplantation and marrow recovery exposes recipients to risks of fatal infection, a significant source of transplant-associated morbidity and mortality. We have previously described the discovery of a small molecule, SW033291, that potently inhibits the prostaglandin degrading enzyme 15-PGDH, increases bone marrow prostaglandin E2, and accelerates hematopoietic recovery following murine transplant. Here we describe the efficacy of (+)-SW209415, a second-generation 15-PGDH inhibitor, in an expanded range of models relevant to human transplantation. (+)-SW209415 is 10,000-fold more soluble, providing the potential for intravenous delivery, while maintaining potency in inhibiting 15-PGDH, increasing in vivo prostaglandin E2, and accelerating hematopoietic regeneration following transplantation. In additional models, (+)-SW209415: (i) demonstrated synergy with granulocyte colony-stimulating factor, the current standard of care; (ii) maintained efficacy as transplant cell dose was escalated; (iii) maintained efficacy when transplant donors and recipients were aged; and (iv) potentiated homing in xenotransplants using human hematopoietic stem cells. (+)-SW209415 showed no adverse effects, no potentiation of in vivo growth of human myeloma and leukemia xenografts, and, on chronic high-dose administration, no toxicity as assessed by weight, blood counts and serum chemistry. These studies provide independent chemical confirmation of the activity of 15-PGDH inhibitors in potentiating hematopoietic recovery, extend the range of models in which inhibiting 15-PGDH demonstrates activity, allay concerns regarding potential for adverse effects from increasing prostaglandin E2, and thereby, advance 15-PGDH as a therapeutic target for potentiating hematopoietic stem cell transplantation.

Collaboration


Dive into the Gretchen A. LaRusch's collaboration.

Top Co-Authors

Avatar

Alvin H. Schmaier

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Evi X. Stavrou

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Fakhri Mahdi

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar

Gregory N. Adams

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Marvin T. Nieman

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Chao Fang

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Yihua Zhou

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mukesh K. Jain

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Alec A. Schmaier

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge