Network


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

Hotspot


Dive into the research topics where Michael R. Landauer is active.

Publication


Featured researches published by Michael R. Landauer.


International Journal of Radiation Biology | 2008

Genistein induces radioprotection by hematopoietic stem cell quiescence

Thomas A. Davis; Ognoon Mungunsukh; Stephen R. Zins; Regina M. Day; Michael R. Landauer

Purpose: In this study we addressed whether genistein-induced radioprotection in mice is associated with alterations of the cell cycle of hematopoietic stem and progenitor cells. Materials and methods: C57BL/6J female mice received a single subcutaneous injection of genistein (200 mg/kg) 24 h prior to a lethal dose (7.75 Gy, 60Co) of total body irradiation. Proliferation-associated Ki-67 protein/7-aminoactinomycin-D (Ki67/7AAD) cell cycle staining was used to differentiate between G0, G1, and S/G2/M in bone marrow cell populations negative for expression of mature hematopoietic lineage marker cells but positive for expression of stem cell antigen-1 and tyrosine kinase receptor for stem cell factor (Lin−Sca-1+cKit+, LSK+). Quantitative real-time polymerase chain reaction (qRT-PCR) microarrays were utilized to examine cell cycle specific genes. Results: At 24 h following radiation exposure, a greater percentage of LSK+ in genistein-treated mice accumulated in the G0 phase of the cell cycle, whereas a large percentage of LSK+ bone marrow cells from untreated and vehicle (PEG-400)-treated mice progressed into the G1 and S/G2/M phases. Moreover, the absolute number of marrow total LSK+, long-term LSK+, and short-term LSK+ increased 2.8, 12.1, and 4.2-fold, respectively, at 7 days post-irradiation in genistein-treated vs. untreated irradiated mice. Lin− cells from genistein-treated mice expressed fewer DNA damage responsive and cell cycle checkpoint genes than LSK+ from untreated or vehicle-treated mice. Conclusion: Pretreatment with genistein provides in vivo protection from acute myelotoxicity through extended quiescence followed by reduced senescence of marrow repopulating LSK+.


International Immunopharmacology | 2009

Effects of genistein administration on cytokine induction in whole-body gamma irradiated mice.

Vijay K. Singh; Marcy B. Grace; Vaishali I. Parekh; Mark H. Whitnall; Michael R. Landauer

The development of an effective pharmacological countermeasure is needed to reduce the morbidity and mortality in military and civilian populations associated with possible exposure to ionizing radiation. We previously demonstrated that a single subcutaneous (sc) administration of genistein at a non-toxic dose provided protection against acute radiation injury and that the radioprotective effects were associated with multilineage, hematopoietic progenitor cell recovery. The purpose of this study was to determine whether hematopoietic recovery was preceded by cytokine induction. In mice treated with sc genistein 24 h before irradiation (7 Gy 60Co), we quantified serum cytokine levels by multiplex Luminex and also investigated a larger number of cytokines using cytokine arrays. Genistein administration stimulated serum granulocyte-colony stimulating factor (G-CSF) 4h and 24h after sham irradiation or gamma-irradiation. Interleukin-6 (IL-6) was significantly increased in genistein-treated animals 4h after irradiation. Because G-CSF and IL-6 are important hematopoietic factors, these results support our hypothesis that the previously observed radioprotective efficacy by genistein may be a result of early recovery of hematopoietic cells due to enhanced production of G-CSF and IL-6.


Experimental Hematology | 2010

Timing of captopril administration determines radiation protection or radiation sensitization in a murine model of total body irradiation

Thomas A. Davis; Michael R. Landauer; Steven Mog; Michal Barshishat-Kupper; Stephen R. Zins; Mihret F. Amare; Regina M. Day

OBJECTIVE Angiotensin II (Ang II), a potent vasoconstrictor, affects the growth and development of hematopoietic cells. Mixed findings have been reported for the effects of angiotensin-converting enzyme (ACE) inhibitors on radiation-induced injury to the hematopoietic system. We investigated the consequences of different regimens of the ACE inhibitor captopril on radiation-induced hematopoietic injury. MATERIALS AND METHODS C57BL/6 mice were either sham-irradiated or exposed to (60)Co total body irradiation (0.6 Gy/min). Captopril was provided in the water for different time periods relative to irradiation. RESULTS In untreated mice, the survival rate from 7.5 Gy was 50% at 30 days postirradiation. Captopril treatment for 7 days prior to irradiation resulted in radiosensitization with 100% lethality and a rapid decline in mature blood cells. In contrast, captopril treatment beginning 1 hour postirradiation and continuing for 30 days resulted in 100% survival, with improved recovery of mature blood cells and multilineage hematopoietic progenitors. In nonirradiated control mice, captopril biphasically modulated Lin(-) marrow progenitor cell cycling. After 2 days, captopril suppressed G(0)-G(1) transition and a greater number of cells entered a quiescent state. However, after 7 days of captopril treatment Lin(-) progenitor cell cycling increased compared to untreated control mice. CONCLUSION These findings suggest that ACE inhibition affects hematopoietic recovery following radiation by modulating the hematopoietic progenitor cell cycle. The timing of captopril treatment relative to radiation exposure differentially affects the viability and repopulation capacity of spared hematopoietic stem cells and, therefore, can result in either radiation protection or radiation sensitization.


Radiation Research | 2013

Genistein Nanoparticles Protect Mouse Hematopoietic System and Prevent Proinflammatory Factors after Gamma Irradiation

Cam T. Ha; Xiang-Hong Li; Dadin Fu; Mang Xiao; Michael R. Landauer

Previous studies demonstrated that genistein protects mice from radiation-induced bone marrow failure. To overcome genisteins extremely low water solubility, a nanoparticle suspension of genistein has been formulated for more rapid dissolution. In the current study, we evaluated the radioprotective effects of a nanoparticle formulation of genistein on survival and hematopoietic recovery in mice exposed to total-body gamma irradiation. A single intramuscular injection of a saline-based genistein nanosuspension (150 mg/kg) administered to CD2F1 mice 24 h before 9.25 Gy 60Co radiation exposure resulted in a 30-day survival rate of 95% compared to 25% in vehicle-treated animals. In mice irradiated at 7 Gy, the genistein nanosuspension increased mouse bone marrow cellularity from approximately 2.9% (vehicle treated) to 28.3% on day 7 postirradiation. Flow cytometry analysis demonstrated decreased radiation-induced hematopoietic stem and progenitor cell (HSPC, Lineage–/cKit+) death from 77.0% (vehicle) to 43.9% (genistein nanosuspension) with a significant recovery of clonogenicity 7 days after irradiation. The genistein nanosuspension also attenuated the radiation-induced elevation of proinflammatory factors interleukin 1 beta (IL-1β), IL-6 and cyclooxygenase-2 (COX-2) in mouse bone marrow and spleen, which may contribute to protecting HSPCs.


PLOS ONE | 2015

Delta-Tocotrienol Suppresses Radiation-Induced MicroRNA-30 and Protects Mice and Human CD34+ Cells from Radiation Injury

Xiang Hong Li; Cam T. Ha; Dadin Fu; Michael R. Landauer; Sanchita P. Ghosh; Mang Xiao

We reported that microRNA-30c (miR-30c) plays a key role in radiation-induced human cell damage through an apoptotic pathway. Herein we further evaluated radiation-induced miR-30 expression and mechanisms of delta-tocotrienol (DT3), a radiation countermeasure candidate, for regulating miR-30 in a mouse model and human hematopoietic CD34+ cells. CD2F1 mice were exposed to 0 (control) or 7–12.5 Gy total-body gamma-radiation, and CD34+ cells were irradiated with 0, 2 or 4 Gy of radiation. Single doses of DT3 (75 mg/kg, subcutaneous injection for mice or 2 μM for CD34+ cell culture) were administrated 24 h before irradiation and animal survival was monitored for 30 days. Mouse bone marrow (BM), jejunum, kidney, liver and serum as well as CD34+ cells were collected at 1, 4, 8, 24, 48 or 72 h after irradiation to determine apoptotic markers, pro-inflammatory cytokines interleukin (IL)-1β and IL-6, miR-30, and stress response protein expression. Our results showed that radiation-induced IL-1β release and cell damage are pathological states that lead to an early expression and secretion of miR-30b and miR-30c in mouse tissues and serum and in human CD34+ cells. DT3 suppressed IL-1β and miR-30 expression, protected against radiation-induced apoptosis in mouse and human cells, and increased survival of irradiated mice. Furthermore, an anti-IL-1β antibody downregulated radiation-induced NFκBp65 phosphorylation, inhibited miR-30 expression and protected CD34+ cells from radiation exposure. Knockdown of NFκBp65 by small interfering RNA (siRNA) significantly suppressed radiation-induced miR-30 expression in CD34+ cells. Our data suggest that DT3 protects human and mouse cells from radiation damage may through suppression of IL-1β-induced NFκB/miR-30 signaling.


Experimental Hematology | 2011

Captopril modulates hypoxia-inducible factors and erythropoietin responses in a murine model of total body irradiation.

Michal Barshishat-Kupper; Ognoon Mungunsukh; Ashlee J. Tipton; Elizabeth A. McCart; Ronald A.M. Panganiban; Thomas A. Davis; Michael R. Landauer; Regina M. Day

OBJECTIVE Our laboratory reported that the angiotensin converting enzyme inhibitor captopril improves erythroid recovery from total body irradiation (TBI) in mice when administered after irradiation. However, captopril administered before TBI attenuates erythroid recovery. Here we investigate captopril and radiation regulation of erythropoietin (EPO) and thrombopoietin (TPO), key effectors of erythroid progenitor proliferation and differentiation. MATERIALS AND METHODS C57BL/6 mice, nonirradiated or exposed to 7.5 Gy TBI ((60)Co, 0.6 Gy/min) were untreated or administered captopril. Plasma EPO and TPO levels were measured by enzyme-linked immunosorbent assay. Gene expression of EPO was determined by quantitative reverse transcription polymerase chain reaction. The hypoxia-inducible factors (HIF)-1α and -2α were measured by immunoblotting. RESULTS In nonirradiated mice, continuous captopril administration in the water transiently reduced reticulocytes and red blood cells after 7 and 10 days, respectively. EPO plasma levels and gene expression were reduced below detectable limits after 2 days of captopril treatment, but recovered within 7 days. HIF-1α and HIF-2α were activated preceding reticulocyte and red blood cell recovery. TBI, which ablates early and late-stage erythroid progenitors, activated both HIFs and increased EPO and TPO. Captopril treatment postirradiation suppressed radiation-induced HIF activation and EPO expression. In contrast, captopril administration for 7 days before TBI resulted in earlier EPO induction and activation. Captopril treatment lowered TPO levels in nonirradiated mice, but had minimal effects on radiation-induced TPO. CONCLUSIONS In nonirradiated mice, captopril biphasically regulates EPO via HIF activation. TBI ablates erythroid progenitors, resulting in hypoxia, HIF activation, and increased EPO expression that are modulated by captopril treatment. These data suggest that short-term suppression of radiation-induced EPO immediately after TBI is favorable for erythroid recovery.


Hormone and Metabolic Research | 2017

Dietary Isoflavone-Dependent and Estradiol Replacement Effects on Body Weight in the Ovariectomized (OVX) Rat

Ashley L. Russell; Jamie Moran Grimes; Danette F. Cruthirds; Joanna Westerfield; Lawren Wooten; Margaret F. Keil; Michael J. Weiser; Michael R. Landauer; Robert J. Handa; T. John Wu; Darwin O. Larco

17β-Estradiol is known to regulate energy metabolism and body weight. Ovariectomy results in body weight gain while estradiol administration results in a reversal of weight gain. Isoflavones, found in rodent chow, can mimic estrogenic effects making it crucial to understand the role of these compounds on metabolic regulation. The goal of this study is to examine the effect of dietary isoflavones on body weight regulation in the ovariectomized rat. This study will examine how dietary isoflavones can interact with estradiol treatment to affect body weight. Consistent with previous findings, animals fed an isoflavone-rich diet had decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin levels (p<0.05) compared to animals fed an isoflavone-free diet. Estradiol replacement resulted in decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin (p<0.05). Current literature suggests the involvement of cytokines in the inflammatory response of body weight gain. We screened a host of cytokines and chemokines that may be altered by dietary isoflavones or estradiol replacement. Serum cytokine analysis revealed significant (p<0.05) diet-dependent increases in inflammatory cytokines (keratinocyte-derived chemokine). The isoflavone-free diet in OVX rats resulted in the regulation of the following cytokines and chemokines: interleukin-10, interleukin-18, serum regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 (p<0.05). Overall, these results reveal that estradiol treatment can have differential effects on energy metabolism and body weight regulation depending on the presence of isoflavones in rodent chow.


Journal of The American Association for Laboratory Animal Science | 2010

Evaluation of Hydration and Nutritional Gels as Supportive Care after Total-Body Irradiation in Mice (Mus musculus)

Krinon D. Moccia; Cara H. Olsen; Jennifer M. Mitchell; Michael R. Landauer


Archive | 2005

Prevention of Gamma Radiation-Induced Mortality in Mice by the Isoflavone Genistein

Michael R. Landauer; V. Srinivasan; M. B. Grace; C. M. Chang; V. Parekh; T. K. Clarke; W. E. Jackson


Journal of Radiation Research | 2017

Accelerated senescence in skin in a murine model of radiation-induced multi-organ injury

Elizabeth A. McCart; Rajesh L. Thangapazham; Eric D. Lombardini; Steven Mog; Ronald Allan M. Panganiban; Kelley M Dickson; Rihab A Mansur; Vitaly Nagy; Sung-Yop Kim; Reed Selwyn; Michael R. Landauer; Thomas N. Darling; Regina M. Day

Collaboration


Dive into the Michael R. Landauer's collaboration.

Top Co-Authors

Avatar

Regina M. Day

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Thomas A. Davis

Naval Medical Research Center

View shared research outputs
Top Co-Authors

Avatar

Cam T. Ha

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Dadin Fu

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. McCart

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Mang Xiao

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Michal Barshishat-Kupper

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Ognoon Mungunsukh

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Stephen R. Zins

Naval Medical Research Center

View shared research outputs
Top Co-Authors

Avatar

Steven Mog

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge