187 results on '"Louis M. Pelus"'
Search Results
2. Further Characterization of Multi-Organ DEARE and Protection by 16,16 Dimethyl Prostaglandin E2 in a Mouse Model of the Hematopoietic Acute Radiation Syndrome
- Author
-
Tong Wu, Louis M. Pelus, P. Artur Plett, Carol H. Sampson, Hui Lin Chua, Alexa Fisher, Hailin Feng, Liqiong Liu, Hongge Li, Miguel Ortiz, Supriya Chittajallu, Qianyi Luo, Ashay D. Bhatwadekar, Timothy B. Meyer, Xin Zhang, Daohong Zhou, Kathryn D. Fischer, David L. McKinzie, Steven J. Miller, and Christie M. Orschell
- Subjects
Radiation ,Biophysics ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
3. Meloxicam with Filgrastim may Reduce Oxidative Stress in Hematopoietic Progenitor Cells during Mobilization of Autologous Peripheral Blood Stem Cells in Patients with Multiple Myeloma
- Author
-
Yunlong Liu, Shuhong Zhang, Andrea M. Patterson, Hongge Li, Sherif S. Farag, Louis M. Pelus, Liqiong Liu, and Pratibha Singh
- Subjects
Filgrastim ,Platelet Engraftment ,CD34 ,Pharmacology ,Meloxicam ,Transplantation, Autologous ,CXCR4 ,Mice ,Autologous stem-cell transplantation ,Heterocyclic Compounds ,medicine ,Animals ,Humans ,business.industry ,Plerixafor ,Hematopoietic Stem Cell Transplantation ,General Medicine ,Hematopoietic Stem Cells ,Hematopoietic Stem Cell Mobilization ,Oxidative Stress ,Peripheral Blood Stem Cells ,Stem cell ,Multiple Myeloma ,business ,medicine.drug - Abstract
Autologous stem cell transplantation (ASCT) is a potentially curative therapy but requires collection of sufficient blood stem cells (PBSC). Up to 40 % of patients with multiple myeloma (MM) fail to collect an optimum number of PBSC using filgrastim only and often require costly plerixafor rescue. The nonsteroidal anti-inflammatory drug meloxicam mobilizes PBSC in mice, nonhuman primates and normal volunteers, and has the potential to attenuate mobilization-induced oxidative stress on stem cells. In a single-center study, we evaluated whether a meloxicam regimen prior to filgrastim increases collection and/or homeostasis of CD34+ cells in MM patients undergoing ASCT. Mobilization was not significantly different with meloxicam in this study; a median of 2.4 × 106 CD34+ cells/kg were collected in the first apheresis and 9.2 × 106 CD34+ cells/kg were collected overall for patients mobilized with meloxicam-filgrastim, versus 4.1 × 106 in first apheresis and 7.2 × 106/kg overall for patients mobilized with filgrastim alone. CXCR4 expression was reduced on CD34+ cells and a higher CD4+/CD8+ T-cell ratio was observed after mobilization with meloxicam-filgrastim. All patients treated with meloxicam-filgrastim underwent ASCT, with neutrophil and platelet engraftment similar to filgrastim alone. RNA sequencing of purified CD34+ cells from 22 MM patients mobilized with meloxicam-filgrastim and 10 patients mobilized with filgrastim only identified > 4,800 differentially expressed genes (FDR
- Published
- 2021
- Full Text
- View/download PDF
4. The Factory of Blood Production: Hematopoietic Stem Cells
- Author
-
Jonathan Hoggatt and Louis M. Pelus
- Published
- 2022
- Full Text
- View/download PDF
5. Hematopoietic Stem Cell Identification Postirradiation
- Author
-
Andrea M. Patterson, Christie M. Orschell, and Louis M. Pelus
- Published
- 2022
- Full Text
- View/download PDF
6. Establishing a Murine Model of the Hematopoietic Acute Radiation Syndrome
- Author
-
P. Artur Plett, Louis M. Pelus, and Christie M. Orschell
- Published
- 2022
- Full Text
- View/download PDF
7. Hematopoietic Stem Cell Identification Postirradiation
- Author
-
Andrea M, Patterson, Christie M, Orschell, and Louis M, Pelus
- Subjects
Mice, Inbred C57BL ,Mice ,Bone Marrow ,Animals ,Bone Marrow Cells ,Hematopoietic Stem Cells ,Hematopoiesis ,Bone Marrow Transplantation - Abstract
Radiation exposure is particularly damaging to cells of the hematopoietic system, inducing pancytopenia and bone marrow failure. The study of these processes, as well as the development of treatments to prevent hematopoietic damage or enhance recovery after radiation exposure, often require analysis of bone marrow cells early after irradiation. While flow cytometry methods are well characterized for identification and analysis of bone marrow populations in the nonirradiated setting, multiple complications arise when dealing with irradiated tissues. Among these complications is a radiation-induced loss of c-Kit, a central marker for conventional gating of primitive hematopoietic populations in mice. These include hematopoietic stem cells (HSCs), which are central to blood reconstitution and life-long bone marrow function, and are important targets of analysis in these studies. This chapter outlines techniques for HSC identification and analysis from mouse bone marrow postirradiation.
- Published
- 2022
8. Establishing a Murine Model of the Hematopoietic Acute Radiation Syndrome
- Author
-
P Artur, Plett, Louis M, Pelus, and Christie M, Orschell
- Subjects
Mice ,Disease Models, Animal ,Acute Radiation Syndrome ,Hematopoietic System ,Animals - Abstract
The hematopoietic system is one of the most sensitive tissues to ionizing radiation, and radiation doses from 2 to 10 gray can result in death from bleeding and infection if left untreated. Reviewing the range of radiation doses reported in the literature that result in similar lethality highlights the need for a more consistent model that would allow a better comparison of the hematopoietic acute radiation syndrome (H-ARS) studies carried out in different laboratories. Developing a murine model of H-ARS to provide a platform suited for efficacy testing of medical countermeasures (MCM) against radiation should include a review of the Food and Drug Administration requirements outlined in the Animal Rule. The various aspects of a murine H-ARS model found to affect consistent performance will be described in this chapter including strain, sex, radiation type and dose, mouse restraint, and husbandry.
- Published
- 2022
9. The magnitude of CXCR4 signaling regulates resistance to quizartinib in FLT3/ITD
- Author
-
Seiji, Fukuda, Nozomi, Matsuda, Tsukimi, Shoji, Chie, Onishi, Tomohiro, Hirade, Takeshi, Taketani, and Louis M, Pelus
- Abstract
CXCR4 antagonists sensitize FLT3/ITD
- Published
- 2022
10. Prostaglandin E2 Regulates Bipotent Monocyte-Dendritic Progenitor Cell Lineage-Commitment
- Author
-
Louis M. Pelus and Pratibha Singh
- Subjects
0301 basic medicine ,biology ,Chemistry ,Monocyte ,General Medicine ,Dendritic cell ,Receptor tyrosine kinase ,Cell biology ,body regions ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,parasitic diseases ,biology.protein ,medicine ,lipids (amino acids, peptides, and proteins) ,Stem cell ,Transcription Factor MafB ,Progenitor cell ,Receptor ,Transcription factor - Abstract
The factors/mechanisms regulating multipotent or bipotent hematopoietic progenitor cells lineage-commitment are not well understood. In this study, we found that prostaglandin E2 (PGE2) is a crucial physiological regulator of lineage choice for the bipotential monocyte-dendritic progenitor cell (MDP). Inhibition of endogenous PGE2 biosynthesis in mice by the dual cyclooxygenase inhibitor, indomethacin, enhances bone marrow and spleen monocyte (MO) differentiation and reduces dendritic cell (DC) differentiation. Ex vivo treatment of purified MDP with indomethacin preferentially increases MO development at the expense of DC generation, whereas addition of exogenous PGE2 reverses the indomethacin-mediated alteration in MDP differentiation potential. Treatment of MDP with selective EP receptor agonists demonstrated that EP1 signaling promotes MDP differentiation into DC at the expense of MO generation. Conversely, EP1 receptor knockout mice showed reduced DC and increased MO differentiation. Mechanistic studies revealed that PGE2 increases expression of the tyrosine kinase receptor Flt3 on MDP and increases the DC-lineage-related transcription factor PU.1, while reducing expression of M-CSFR and the MO-lineage-related transcription factor MafB. These data indicate that PGE2-EP1 signaling plays a critical role in MDP lineage commitment and DC and MO differentiation.
- Published
- 2021
- Full Text
- View/download PDF
11. Prostaglandin E2 Enhances Aged Hematopoietic Stem Cell Function
- Author
-
P. Artur Plett, Yunlong Liu, Christie M. Orschell, Louis M. Pelus, Carol H. Sampson, Edward Simpson, and Andrea M. Patterson
- Subjects
0301 basic medicine ,Myeloid ,Prostaglandin E2 receptor ,Hematopoietic stem cell ,hemic and immune systems ,Biology ,Transplantation ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Bone marrow ,Stem cell ,Prostaglandin E2 ,medicine.drug - Abstract
Aging of hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSCs), along with functional compromise and myeloid bias, with donor age being a significant variable in survival after HSC transplantation. No clinical methods currently exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSCs to biological stimuli. Exposure of HSCs from young fish, mice, nonhuman primates, and humans to 16,16-dimethyl prostaglandin E2 (dmPGE2) enhances transplantation, but the effect of dmPGE2 on aged HSCs is unknown. Here we show that ex vivo pulse of bone marrow cells from young adult (3 mo) and aged (25 mo) mice with dmPGE2 prior to serial competitive transplantation significantly enhanced long-term repopulation from aged grafts in primary and secondary transplantation (27 % increase in chimerism) to a similar degree as young grafts (21 % increase in chimerism; both p
- Published
- 2021
- Full Text
- View/download PDF
12. Hematopoietic Agents
- Author
-
Jonathan Hoggatt, Shruti Datari, and Louis M. Pelus
- Published
- 2021
- Full Text
- View/download PDF
13. A Single Radioprotective Dose of Prostaglandin E2 Blocks Irradiation-Induced Apoptotic Signaling and Early Cycling of Hematopoietic Stem Cells
- Author
-
Jonathan Hoggatt, Khalid S. Mohammad, Andrea M. Patterson, Carol H. Sampson, Liqiong Liu, Maegan L. Capitano, Hongge Li, P. Artur Plett, Pratibha Singh, Christie M. Orschell, and Louis M. Pelus
- Subjects
0301 basic medicine ,Hematopoietic stem cell ,Cell Biology ,Biology ,Cell cycle ,Biochemistry ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Bone marrow suppression ,Genetics ,medicine ,Cancer research ,Bone marrow ,Stem cell ,Prostaglandin E2 ,Progenitor cell ,030217 neurology & neurosurgery ,Developmental Biology ,medicine.drug - Abstract
Summary Ionizing radiation exposure results in acute and delayed bone marrow suppression. Treatment of mice with 16,16-dimethyl prostaglandin E2 (dmPGE2) prior to lethal ionizing radiation (IR) facilitates survival, but the cellular and molecular mechanisms are unclear. In this study we show that dmPGE2 attenuates loss and enhances recovery of bone marrow cellularity, corresponding to a less severe hematopoietic stem cell nadir, and significantly preserves long-term repopulation capacity and progenitor cell function. Mechanistically, dmPGE2 suppressed hematopoietic stem cell (HSC) proliferation through 24 h post IR, which correlated with fewer DNA double-strand breaks and attenuation of apoptosis, mitochondrial compromise, oxidative stress, and senescence. RNA sequencing of HSCs at 1 h and 24 h post IR identified a predominant interference with IR-induced p53-downstream gene expression at 1 h, and confirmed the suppression of IR-induced cell-cycle genes at 24 h. These data identify mechanisms of dmPGE2 radioprotection and its potential role as a medical countermeasure against radiation exposure.
- Published
- 2020
- Full Text
- View/download PDF
14. CXCR4 expression in the bone marrow microenvironment is required for hematopoietic stem and progenitor cell maintenance and early hematopoietic regeneration after myeloablation
- Author
-
Khalid S. Mohammad, Pratibha Singh, and Louis M. Pelus
- Subjects
0301 basic medicine ,Receptors, CXCR4 ,Transplantation Conditioning ,Stromal cell ,Bone Marrow Cells ,Stem cell factor ,Biology ,CXCR4 ,Article ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,medicine ,Animals ,Progenitor cell ,Mesenchymal stem cell ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Molecular Medicine ,Bone marrow ,030217 neurology & neurosurgery ,Developmental Biology ,Homing (hematopoietic) - Abstract
The bone marrow (BM) microenvironment/niche plays a key role in regulating hematopoietic stem and progenitor cell (HSPC) activities; however, mechanisms regulating niche cell function are not well understood. In this study, we show that niche intrinsic expression of the CXCR4 chemokine receptor critically regulates HSPC maintenance during steady state, and promotes early hematopoietic regeneration after myeloablative irradiation. At steady state, chimeric mice with wild-type (WT) HSPC and marrow stroma that lack CXCR4 show decreased HSPC quiescence, and their repopulation capacity was markedly reduced. Mesenchymal stromal cells (MSC) were significantly reduced in the BM of CXCR4 deficient mice, which was accompanied by decreased levels of the HSPC supporting factors stromal cell-derived factor-1 (SDF-1) and stem cell factor (SCF). CXCR4 also plays a crucial role in survival and restoration of BM stromal cells after myeloablative irradiation, where the loss of BM stromal cells was more severe in CXCR4-deficient mice compared to WT mice. In addition, transplantation of WT donor HSPC into CXCR4-deficient recipient mice demonstrated reduced HSPC homing and early hematopoietic reconstitution. We found that CXCR4 signaling attenuates irradiation-induced BM stromal cell loss by upregulating the expression of the antiapoptotic protein Survivin via the PI3K pathway. Our study suggests that SDF-1-CXCR4 signaling in the stromal microenvironment cells plays a crucial role in maintenance of HSPCs during homeostasis, and promotes niche regeneration and early hematopoietic reconstitution after transplantation. Modulation of CXCR4 signaling in the HSPC microenvironment could be a means to enhance hematopoietic recovery after clinical hematopoietic cell transplantation.
- Published
- 2020
- Full Text
- View/download PDF
15. Age and Sex Divergence in Hematopoietic Radiosensitivity in Aged Mouse Models of the Hematopoietic Acute Radiation Syndrome
- Author
-
Andrea M. Patterson, Sasidhar Vemula, P. Artur Plett, Carol H. Sampson, Hui Lin Chua, Alexa Fisher, Tong Wu, Rajendran Sellamuthu, Hailin Feng, Barry P. Katz, Colleen M. DesRosiers, Louis M. Pelus, George N. Cox, Thomas J. MacVittie, and Christie M. Orschell
- Subjects
Male ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Radiation ,Acute Radiation Syndrome ,Biophysics ,Animals ,Radiology, Nuclear Medicine and imaging ,Female ,Hematopoietic Stem Cells ,Radiation Tolerance ,Hematopoiesis - Abstract
The hematopoietic system is highly sensitive to stress from both aging and radiation exposure, and the hematopoietic acute radiation syndrome (H-ARS) should be modeled in the geriatric context separately from young for development of age-appropriate medical countermeasures (MCMs). Here we developed aging murine H-ARS models, defining radiation dose response relationships (DRRs) in 12-month-old middle-aged and 24-month-old geriatric male and female C57BL/6J mice, and characterized diverse factors affecting geriatric MCM testing. Groups of approximately 20 mice were exposed to ∼10 different doses of radiation to establish radiation DRRs for estimation of the LD50/30. Radioresistance increased with age and diverged dramatically between sexes. The LD50/30 in young adult mice averaged 853 cGy and was similar between sexes, but increased in middle age to 1,005 cGy in males and 920 cGy in females, with further sex divergence in geriatric mice to 1,008 cGy in males but 842 cGy in females. Correspondingly, neutrophils, platelets, and functional hematopoietic progenitor cells were all increased with age and rebounded faster after irradiation. These effects were higher in aged males, and neutrophil dysfunction was observed in aged females. Upstream of blood production, hematopoietic stem cell (HSC) markers associated with age and myeloid bias (CD61 and CD150) were higher in geriatric males vs. females, and sex-divergent gene signatures were found in HSCs relating to cholesterol metabolism, interferon signaling, and GIMAP family members. Fluid intake per gram body weight decreased with age in males, and decreased after irradiation in all mice. Geriatric mice of substrain C57BL/6JN sourced from the National Institute on Aging were significantly more radiosensitive than C57BL/6J mice from Jackson Labs aged at our institution, indicating mouse source and substrain should be considered in geriatric radiation studies. This work highlights the importance of sex, vendor, and other considerations in studies relating to hematopoiesis and aging, identifies novel sex-specific functional and molecular changes in aging hematopoietic cells at steady state and after irradiation, and presents well-characterized aging mouse models poised for MCM efficacy testing for treatment of acute radiation effects in the elderly.
- Published
- 2022
16. Upregulation of SIRT1 Contributes to dmPGE2-dependent Radioprotection of Hematopoietic Stem Cells
- Author
-
Liqiong, Liu, Hongge, Li, Andrea M, Patterson, P Artur, Plett, Carol H, Sampson, Khalid S, Mohammad, Maegan L, Capitano, Pratibha, Singh, Chonghua, Yao, Christie M, Orschell, and Louis M, Pelus
- Subjects
Mice ,Sirtuin 1 ,Animals ,Apoptosis ,Tumor Suppressor Protein p53 ,Hematopoietic Stem Cells ,Up-Regulation - Abstract
Exposure to potentially lethal high-dose ionizing radiation results in bone marrow suppression, known as the hematopoietic acute radiation syndrome (H-ARS), which can lead to pancytopenia and possible death from hemorrhage or infection. Medical countermeasures to protect from or mitigate the effects of radiation exposure are an ongoing medical need. We recently reported that 16,16 dimethyl prostaglandin E
- Published
- 2022
17. The magnitude of CXCR4 signaling regulates resistance to quizartinib in FLT3/ITD+ cells via RUNX1
- Author
-
Seiji Fukuda, Nozomi Matsuda, Tsukimi Shoji, Chie Onishi, Tomohiro Hirade, Takeshi Taketani, and Louis M. Pelus
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
- Full Text
- View/download PDF
18. Aging negatively impacts the ability of megakaryocytes to stimulate osteoblast proliferation and bone mass
- Author
-
Evan R. Himes, Kevin A. Maupin, Joydeep Ghosh, Louis M. Pelus, Christie M. Orschell, Paul Childress, Alexa Fisher, Marta B. Alvarez, Jung Min Hong, Irushi Abeysekera, Edward F. Srour, Pratibha Singh, Artur P. Plett, Safa F. Mohamad, Carol H. Sampson, Melissa A. Kacena, Angela Bruzzaniti, and Hui Lin Chua
- Subjects
Male ,0301 basic medicine ,Aging ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Cell Count ,030209 endocrinology & metabolism ,Article ,Bone and Bones ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Bone Marrow ,Internal medicine ,medicine ,Animals ,GATA1 Transcription Factor ,Bone formation ,Cell Proliferation ,Osteoblasts ,biology ,urogenital system ,Chemistry ,Osteoblast ,Organ Size ,X-Ray Microtomography ,Transforming growth factor beta ,Hematopoietic Stem Cells ,Adoptive Transfer ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,Phenotype ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,biology.protein ,Megakaryocytes ,Bone mass - Abstract
Osteoblast number and activity decreases with aging, contributing to the age-associated decline of bone mass, but the mechanisms underlying changes in osteoblast activity are not well understood. Here, we show that the age-associated bone loss critically depends on impairment of the ability of megakaryocytes (MKs) to support osteoblast proliferation. Co-culture of osteoblast precursors with young MKs is known to increase osteoblast proliferation and bone formation. However, co-culture of osteoblast precursors with aged MKs resulted in significantly fewer osteoblasts compared to co-culture with young MKs, and this was associated with the downregulation of transforming growth factor beta. In addition, the ability of MKs to increase bone mass was attenuated during aging as transplantation of GATA1(low/low) hematopoietic donor cells (which have elevated MKs/MK precursors) from young mice resulted in an increase in bone mass of recipient mice compared to transplantation of young wild-type donor cells, whereas transplantation of GATA1(low/low) donor cells from old mice failed to enhance bone mass in recipient mice compared to transplantation of old wild-type donor cells. These findings suggest that the preservation or restoration of the MK-mediated induction of osteoblast proliferation during aging may hold the potential to prevent age-associated bone loss and resulting fractures.
- Published
- 2019
- Full Text
- View/download PDF
19. Prostaglandin E
- Author
-
Pratibha, Singh and Louis M, Pelus
- Subjects
Mice ,Animals ,Cell Differentiation ,Hematopoietic Stem Cells ,Dinoprostone ,Monocytes ,Signal Transduction - Abstract
The factors/mechanisms regulating multipotent or bipotent hematopoietic progenitor cells lineage-commitment are not well understood. In this study, we found that prostaglandin E
- Published
- 2021
20. Prostaglandin E
- Author
-
Andrea M, Patterson, P Artur, Plett, Carol H, Sampson, Edward, Simpson, Yunlong, Liu, Louis M, Pelus, and Christie M, Orschell
- Subjects
Mice ,Prostaglandins E ,Prostaglandins ,Animals ,RNA, Messenger ,Hematopoietic Stem Cells - Abstract
Aging of hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSCs), along with functional compromise and myeloid bias, with donor age being a significant variable in survival after HSC transplantation. No clinical methods currently exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSCs to biological stimuli. Exposure of HSCs from young fish, mice, nonhuman primates, and humans to 16,16-dimethyl prostaglandin E
- Published
- 2021
21. Prostaglandin E2 Enhances Aged Hematopoietic Stem Cell Function
- Author
-
Andrea M. Patterson, P. Artur Plett, Carol H. Sampson, Edward Simpson, Yunlong Liu, Louis M. Pelus, and Christie M. Orschell
- Abstract
Aging of hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSCs), along with functional compromise and myeloid bias, with donor age being a significant variable in survival after HSC transplantation. No clinical methods currently exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSCs to biological stimuli. Exposure of HSCs from young fish, mice, nonhuman primates, and humans to 16,16-dimethyl prostaglandin E2 (dmPGE2) enhances transplantation, but the effect of dmPGE2 on aged HSCs is unknown. Here we show that ex vivo pulse of bone marrow cells from young adult (3 mo) and aged (25 mo) mice with dmPGE2 prior to serial competitive transplantation significantly enhanced long-term repopulation from aged grafts in primary and secondary transplantation (27% increase in chimerism) to a similar degree as young grafts (21% increase in chimerism; both p2 are similar in young and old, including CREB1 activation and increased cell survival and homeostasis. Common genes within these pathways identified likely key mediators of HSC enhancement by dmPGE2 and age-related signaling differences. HSC expression of the PGE2 receptor EP4, implicated in HSC function, increased with age in both mRNA and surface protein. This work suggests that aging does not alter the major dmPGE2 response pathways in HSCs which mediate enhancement of both young and old HSC function, with significant implications for expanding the therapeutic potential of elderly HSC transplantation.
- Published
- 2021
- Full Text
- View/download PDF
22. Optimizing and Profiling Prostaglandin E2 as a Medical Countermeasure for the Hematopoietic Acute Radiation Syndrome
- Author
-
Hui Lin Chua, Andrea M. Patterson, Carol H. Sampson, P. Artur Plett, Jessica L. Muldoon, Christie M. Orschell, Hailin Feng, Tong Wu, Alexa Fisher, Pratibha Singh, Theresa A. Guise, Laura E. Wright, and Louis M. Pelus
- Subjects
Prostaglandin E2 receptor ,Biophysics ,Radiation-Protective Agents ,Pharmacology ,Radiation Tolerance ,Dinoprostone ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Peripheral blood cell ,Prostaglandin E2 ,Radiation ,business.industry ,Interleukin-6 ,Sequence Analysis, RNA ,Mesenchymal stem cell ,Acute Radiation Syndrome ,Dose-Response Relationship, Radiation ,Hematopoietic Stem Cells ,Hematopoiesis ,Haematopoiesis ,medicine.anatomical_structure ,Gene Expression Regulation ,Gamma Rays ,030220 oncology & carcinogenesis ,Bone marrow ,Stem cell ,business ,Whole-Body Irradiation ,medicine.drug - Abstract
Identification of medical countermeasures (MCM) to mitigate radiation damage and/or protect first responders is a compelling unmet medical need. The prostaglandin E(2) (PGE(2)) analog, 16,16 dimethyl-PGE(2) (dmPGE(2)), has shown efficacy as a radioprotectant and radiomitigator that can enhance hematopoiesis and ameliorate intestinal mucosal cell damage. In this study, we optimized the time of administration of dmPGE(2) for protection and mitigation against mortality from the hematopoietic acute radiation syndrome (H-ARS) in young adult mice, evaluated its activity in pediatric and geriatric populations, and investigated potential mechanisms of action. Windows of 30-day survival efficacy for single administration of dmPGE(2) were defined as within 3 h prior to and 6–30 h after total-body γ irradiation (TBI). Radioprotective and radio-mitigating efficacy was also observed in 2-year-old geriatric mice and 6-week-old pediatric mice. PGE(2) receptor agonist studies suggest that signaling through EP4 is primarily responsible for the radioprotective effects. DmPGE(2) administration prior to TBI attenuated the drop in red blood cells and platelets, accelerated recovery of all peripheral blood cell types, and resulted in higher hematopoietic and mesenchymal stem cells in survivor bone marrow. Multiplex analysis of bone marrow cytokines together with RNA sequencing of hematopoietic stem cells indicated a pro-hematopoiesis cytokine milieu induced by dmPGE(2), with IL-6 and G-CSF strongly implicated in dmPGE(2)-mediated radioprotective activity. In summary, we have identified windows of administration for significant radio-mitigation and radioprotection by dmPGE(2) in H-ARS, demonstrated survival efficacy in special populations, and gained insight into radioprotective mechanisms, information useful towards development of dmPGE(2) as a MCM for first responders, military personnel, and civilians facing radiation threats.
- Published
- 2020
23. Aging-Related Reduced Expression of CXCR4 on Bone Marrow Mesenchymal Stromal Cells Contributes to Hematopoietic Stem and Progenitor Cell Defects
- Author
-
Christie M. Orschell, Pratibha Singh, Melissa A. Kacena, and Louis M. Pelus
- Subjects
0301 basic medicine ,Premature aging ,Senescence ,Aging ,Receptors, CXCR4 ,Cell Count ,Biology ,CXCR4 ,Article ,Colony-Forming Units Assay ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Progenitor cell ,Cells, Cultured ,Mesenchymal stem cell ,Cell Cycle ,Mesenchymal Stem Cells ,Free Radical Scavengers ,Hematopoietic Stem Cells ,Cell biology ,Acetylcysteine ,Clone Cells ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,030220 oncology & carcinogenesis ,Bone marrow ,Stem cell ,Reactive Oxygen Species - Abstract
Aging impairs the regenerative potential of hematopoietic stem cells (HSC) and skews differentiation towards the myeloid lineage. The bone marrow (BM) microenvironment has recently been suggested to influence HSC aging, however the mechanisms whereby BM stromal cells mediate this effect is unknown. Here we show that aging-associated decreased expression of CXCR4 expression on BM mesenchymal stem cells (MSC) plays a crucial role in the development of the hematopoietic stem and progenitor cells (HSPC) aging phenotype. The BM MSC from old mice was sufficient to drive a premature aging phenotype of young HSPC when cultured together ex vivo. The impaired ability of old MSC to support HSPC function is associated with reduced expression of CXCR4 on BM MSC of old mice. Deletion of the CXCR4 gene in young MSC accelerates an aging phenotype in these cells characterized by increased production of reactive oxygen species (ROS), DNA damage, senescence, and reduced proliferation. Culture of HSPC from young mice with CXCR4 deficient MSC also from young mice led to a premature aging phenotype in the young HSPC, as evidenced by reduced hematopoietic regeneration and enhanced myeloid differentiation. Mechanistically, CXCR4 signaling prevents BM MSC dysfunction by suppressing oxidative stress, as treatment of old or CXCR4 deficient MSC with N-acetyl-L-cysteine (NAC), improved their niche supporting activity, and attenuated the HSPC aging phenotype. Our studies suggest that age-associated reduction in CXCR4 expression on BM MSC impairs hematopoietic niche activity with increased ROS production, driving an HSC aging phenotype. Thus, modulation of the SDF-1/CXCR4 axis in MSC may lead to novel interventions to alleviate the age-associated decline in immune/hematopoietic function.
- Published
- 2020
24. Obituary for Dr. Hal Broxmeyer
- Author
-
Louis M. Pelus
- Published
- 2022
- Full Text
- View/download PDF
25. Neuropeptide Y regulates a vascular gateway for hematopoietic stem and progenitor cells
- Author
-
Malgorzata M. Kamocka, Khalid S. Mohammad, Nadia Carlesso, Jonathan Hoggatt, Mary R. Saunders, Louis M. Pelus, Pratibha Singh, Theresa A. Guise, Jennifer M. Speth, and Hongge Li
- Subjects
0301 basic medicine ,Dipeptidyl Peptidase 4 ,Vascular permeability ,Mice ,03 medical and health sciences ,mental disorders ,Animals ,Humans ,Neuropeptide Y ,Progenitor cell ,Receptor ,Mice, Knockout ,Chemistry ,Endothelial Cells ,General Medicine ,Hematopoietic Stem Cells ,Neuropeptide Y receptor ,humanities ,Receptors, Neuropeptide Y ,Cell biology ,Endothelial stem cell ,Transplantation ,Haematopoiesis ,030104 developmental biology ,Commentary ,Stem cell ,Signal Transduction - Abstract
Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.
- Published
- 2017
- Full Text
- View/download PDF
26. A Single Radioprotective Dose of Prostaglandin E
- Author
-
Andrea M, Patterson, Liqiong, Liu, Carol H, Sampson, P Artur, Plett, Hongge, Li, Pratibha, Singh, Khalid S, Mohammad, Jonathan, Hoggatt, Maegan L, Capitano, Christie M, Orschell, and Louis M, Pelus
- Subjects
p53 ,hematopoietic regeneration ,bone marrow ,Transcription, Genetic ,Apoptosis ,Radiation-Protective Agents ,Dinoprostone ,Article ,stem cells ,Radiation, Ionizing ,Animals ,Gene Regulatory Networks ,16,16-dimethyl PGE2 ,Cell Cycle ,RNA sequencing ,Hematopoietic Stem Cells ,Hematopoiesis ,Mice, Inbred C57BL ,radiation ,Gene Expression Regulation ,DNA damage ,cell cycle ,prostaglandin ,DNA Damage ,Signal Transduction ,Transcription Factors - Abstract
Summary Ionizing radiation exposure results in acute and delayed bone marrow suppression. Treatment of mice with 16,16-dimethyl prostaglandin E2 (dmPGE2) prior to lethal ionizing radiation (IR) facilitates survival, but the cellular and molecular mechanisms are unclear. In this study we show that dmPGE2 attenuates loss and enhances recovery of bone marrow cellularity, corresponding to a less severe hematopoietic stem cell nadir, and significantly preserves long-term repopulation capacity and progenitor cell function. Mechanistically, dmPGE2 suppressed hematopoietic stem cell (HSC) proliferation through 24 h post IR, which correlated with fewer DNA double-strand breaks and attenuation of apoptosis, mitochondrial compromise, oxidative stress, and senescence. RNA sequencing of HSCs at 1 h and 24 h post IR identified a predominant interference with IR-induced p53-downstream gene expression at 1 h, and confirmed the suppression of IR-induced cell-cycle genes at 24 h. These data identify mechanisms of dmPGE2 radioprotection and its potential role as a medical countermeasure against radiation exposure., Graphical Abstract, Highlights • Treatment with dmPGE2 prior to lethal IR attenuates early hematopoietic cell loss • IR-induced apoptotic gene expression is blocked in HSCs by dmPGE2 • dmPGE2 temporarily suppresses proliferation of HSCs and HPCs in vivo • Early IR-induced cycling, DNA damage, and their sequelae were attenuated in HSCs, In this article, Pelus, Orschell, and colleagues demonstrate that dmPGE2 protection from lethal radiation preserves HSC numbers and repopulating capacity, which is associated with attenuation of HSC cycling and DNA damage accumulation within the first critical day. Genomically, dmPGE2 predominantly blocks HSC induction of p53-regulated apoptotic genes within 1 h of irradiation, altogether promoting hematopoietic recovery and survival.
- Published
- 2020
27. Cardiac and Renal Delayed Effects of Acute Radiation Exposure: Organ Differences in Vasculopathy, Inflammation, Senescence and Oxidative Balance
- Author
-
Artur Plett, Miguel A Ortiz, Joseph L. Unthank, Carol H. Sampson, Louis M. Pelus, Steven J. Miller, Hina Trivedi, Eric P. Cohen, Christie M. Orschell, Hui Lin Chua, Rajendran Sellamuthu, and Alexa Fisher
- Subjects
Senescence ,Male ,Pathology ,medicine.medical_specialty ,Time Factors ,Biophysics ,Inflammation ,Cell Count ,medicine.disease_cause ,Kidney ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adventitia ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Cellular Senescence ,Radiation ,business.industry ,Myocardium ,Kidney metabolism ,Dose-Response Relationship, Radiation ,Heart ,Coronary arteries ,Mice, Inbred C57BL ,Dose–response relationship ,Oxidative Stress ,medicine.anatomical_structure ,Acute Radiation Syndrome ,Organ Specificity ,030220 oncology & carcinogenesis ,Disease Progression ,Blood Vessels ,Female ,medicine.symptom ,business ,Oxidative stress ,Whole-Body Irradiation - Abstract
We have previously shown significant pathology in the heart and kidney of murine hematopoietic-acute radiation syndrome (H-ARS) survivors of 8.7-9.0 Gy total-body irradiation (TBI). The goal of this study was to determine temporal relationships in the development of vasculopathy and the progression of renal and cardiovascular delayed effects of acute radiation exposure (DEARE) at TBI doses less than 9 Gy and to elucidate the potential roles of senescence, inflammation and oxidative stress. Our results show significant loss of endothelial cells in coronary arteries by 4 months post-TBI (8.53 or 8.72 Gy of gamma radiation). This loss precedes renal dysfunction and interstitial fibrosis and progresses to abnormalities in the arterial media and adventitia and loss of coronary arterioles. Major differences in radiation-induced pathobiology exist between the heart and kidney in terms of vasculopathy progression and also in indices of inflammation, senescence and oxidative imbalance. The results of this work suggest a need for different medical countermeasures for multiple targets in different organs and at various times after acute radiation injury to prevent the progression of DEARE.
- Published
- 2019
28. 3028 – ENHANCING AGED HEMATOPOIETIC STEM CELL FUNCTION WITH PROSTAGLANDIN E2
- Author
-
Christie M. Orschell, Carol H. Sampson, Andrea M. Patterson, Louis M. Pelus, and P. Artur Plett
- Subjects
Cancer Research ,Myeloid ,business.industry ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Transplantation ,Andrology ,Haematopoiesis ,medicine.anatomical_structure ,Genetics ,medicine ,Bone marrow ,Stem cell ,Prostaglandin E2 ,business ,Molecular Biology ,Ex vivo ,medicine.drug - Abstract
Aging of human hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSC), along with functional compromise and myeloid bias, and donor age is a significant variable in survival after HSC transplantation. No clinical methods exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSC to biological stimuli. Exposure of young fish, mouse, nonhuman primate and human HSC to 16,16-dimethyl prostaglandin E2 (dmPGE2) can enhance transplantation, but the effect of dmPGE2 on aged HSC has not yet been tested. Bone marrow cells from young (3 mo) and aged (25 mo) C57BL/6J mice were pulsed ex vivo with dmPGE2 or vehicle prior to competitive transplantation. DmPGE2 enhanced long-term repopulation of aged grafts in primary and secondary transplantation, increasing final chimerism an average of 27%, similar to the average increase of 21% for young grafts (both p
- Published
- 2020
- Full Text
- View/download PDF
29. Peripheral blood stem cell mobilization; a look ahead
- Author
-
Hal E. Broxmeyer and Louis M. Pelus
- Subjects
0301 basic medicine ,Mobilization ,Genetic enhancement ,Cell Biology ,Biology ,Peripheral blood ,Article ,Transplantation ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,Genetics ,Stem cell ,Progenitor cell ,Molecular Biology ,Neuroscience ,Hematopoietic Stem Cell Mobilization ,Developmental Biology - Abstract
THE PURPOSE OF REVIEW: Mobilized peripheral blood is the predominant source of stem and progenitor cells for hematologic transplantation. Successful transplant requires sufficient stem cells of high enough quality to recapitulate lifelong hematopoiesis, but in some patients and normal donors, reaching critical threshold stem cell numbers are difficult to achieve. Novel strategies, particularly those offering rapid mobilization and reduced costs, remains an area of interest. This review summarizes critical scientific underpinnings in understanding the process of stem cell mobilization, with a focus on new or improved strategies for their efficient collection and engraftment. RECENT FINDINGS: Studies are described that provide new insights into the complexity of stem cell mobilization. Agents that target new pathways such HSC egress, identify strategies to collect more potent competing HSC and new methods to optimize stem cell collection and engraftment are being evaluated. SUMMARY: Agents and more effective strategies that directly address the current shortcomings of hematopoietic stem cell mobilization and transplantation and offer the potential to facilitate collection and expand use of mobilized stem cells have been identified.
- Published
- 2018
30. Role of lipegfilgrastim in the management of chemotherapy-induced neutropenia
- Author
-
Tiffany A. Tate, Louis M. Pelus, and Jonathan Hoggatt
- Subjects
medicine.medical_specialty ,Letter ,Opportunistic infection ,medicine.medical_treatment ,XM22 ,Biophysics ,Pharmaceutical Science ,Bioengineering ,Review ,granulocyte-colony stimulating factor ,Neutropenia ,Filgrastim ,Polyethylene Glycols ,Biomaterials ,Drug Discovery ,Granulocyte Colony-Stimulating Factor ,medicine ,Humans ,cancer ,Chemotherapy-Induced Febrile Neutropenia ,Intensive care medicine ,Chemotherapy ,business.industry ,Organic Chemistry ,neutrophil ,Biosimilar ,General Medicine ,medicine.disease ,Recombinant Proteins ,Granulocyte colony-stimulating factor ,biosimilar ,business ,Lipegfilgrastim ,filgrastim ,Pegfilgrastim ,medicine.drug - Abstract
Chemotherapy, irradiation, and other agents are widely used to target the process of cell division in neoplastic cells. However, while these therapies are effective against most cancers, the high proliferative rate of the cells of the hematopoietic system that produce billions of blood cells needed daily throughout life is extremely sensitive to these agents, resulting in loss of blood cell populations, which can be life threatening. Neutropenia is the most serious hematologic toxicity of chemotherapy, which can result in patient morbidity and mortality due to opportunistic infection and often is the limiting factor in dose escalation or duration of chemotherapeutic administration. Neutropenic patients often require hospitalization and incur substantial medical costs associated with anti-infective therapy. Treatment of iatrogenic and congenic neutropenia was changed in the early 1990s with the introduction of filgrastim (Neupogen(®)) and pegfilgrastim (Neulasta(®)). With the expiration of patent lives of both of these drugs, biosimilars have begun to emerge. In this review, we will summarize the chemical characteristics, pharmacokinetics, safety and efficacy of lipegfilgrastim (Lonquex(®)), the first long-acting biosimilar filgrastim to receive regulatory approval and enter the marketplace.
- Published
- 2015
31. G-CSF in stem cell mobilization: new insights, new questions
- Author
-
Louis M. Pelus and Andrea M. Patterson
- Subjects
0301 basic medicine ,Platelet Membrane Glycoprotein IIb ,Stem cell mobilization ,Mice, Transgenic ,Biology ,Article ,03 medical and health sciences ,Mice ,Lineage tracing ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Cell Proliferation ,Mobilization ,hemic and immune systems ,Hematology ,Hematopoietic Stem Cells ,Phenotype ,Peripheral blood ,Hematopoietic Stem Cell Mobilization ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunology ,Bone marrow ,Stem cell - Abstract
Granulocyte colony-stimulating factor (G-CSF) is used clinically to treat leukopenia and to enforce hematopoietic stem cell (HSC) mobilization to the peripheral blood (PB). However, G-CSF is also produced in response to infection, and excessive exposure reduces HSC repopulation capacity. Previous work has shown that dormant HSCs contain all the long-term repopulation potential in the bone marrow (BM), and that as HSCs accumulate a divisional history, they progressively lose regenerative potential. As G-CSF treatment also induces HSC proliferation, we sought to examine whether G-CSF-mediated repopulation defects are a result of increased proliferative history. To do so, we used an established H2BGFP label retaining system to track HSC divisions in response to G-CSF. Our results show that dormant HSCs are preferentially mobilized to the PB on G-CSF treatment. We find that this mobilization does not result in H2BGFP label dilution of dormant HSCs, suggesting that G-CSF does not stimulate dormant HSC proliferation. Instead, we find that proliferation within the HSC compartment is restricted to CD41-expressing cells that function with short-term, and primarily myeloid, regenerative potential. Finally, we show CD41 expression is up-regulated within the BM HSC compartment in response to G-CSF treatment. This emergent CD41
- Published
- 2017
32. Survivin modulates genes with divergent molecular functions and regulates proliferation of hematopoietic stem cells through Evi-1
- Author
-
Jennifer M. Speth, Jonathan Hoggatt, Pratibha Singh, Louis M. Pelus, Peirong Hu, Mariko Abe, Seiji Yamaguchi, Giuseppina Nucifora, Seiji Fukuda, and Edward M. Conway
- Subjects
Cancer Research ,Transcription, Genetic ,Survivin ,Bone Marrow Cells ,Biology ,Inhibitor of apoptosis ,Article ,Inhibitor of Apoptosis Proteins ,Fusion gene ,Mice ,Proto-Oncogenes ,Animals ,Humans ,neoplasms ,Transcription factor ,Alleles ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Homeodomain Proteins ,Regulation of gene expression ,Gene Expression Regulation, Leukemic ,Cell Cycle ,Pre-B-Cell Leukemia Transcription Factor 1 ,GATA2 ,Intracellular Signaling Peptides and Proteins ,hemic and immune systems ,Hematology ,Hematopoietic Stem Cells ,MDS1 and EVI1 Complex Locus Protein ,Hematopoiesis ,DNA-Binding Proteins ,GATA2 Transcription Factor ,Mice, Inbred C57BL ,Repressor Proteins ,Haematopoiesis ,Phenotype ,Retroviridae ,Oncology ,Cancer research ,Female ,Stem cell ,Gene Deletion ,Transcription Factors - Abstract
The inhibitor of apoptosis protein Survivin regulates hematopoiesis, although its mechanisms of regulation of hematopoietic stem cells (HSCs) remain largely unknown. While investigating conditional Survivin deletion in mice, we found that Survivin was highly expressed in phenotypically defined HSCs, and Survivin deletion in mice resulted in significantly reduced total marrow HSCs and hematopoietic progenitor cells. Transcriptional analysis of Survivin(-/-) HSCs revealed altered expression of multiple genes not previously linked to Survivin activity. In particular, Survivin deletion significantly reduced expression of the Evi-1 transcription factor indispensable for HSC function, and the downstream Evi-1 target genes Gata2, Pbx1 and Sall2. The loss of HSCs following Survivin deletion and impaired long-term HSC repopulating function could be partially rescued by ectopic Evi-1 expression in Survivin -/- HSCs. These data demonstrate that Survivin partially regulates HSC function by modulating the Evi-1 transcription factor and its downstream targets and identify new genetic pathways in HSCs regulated by Survivin.
- Published
- 2014
- Full Text
- View/download PDF
33. Inhibition of DPP4/CD26 and dmPGE2 treatment enhances engraftment of mouse bone marrow hematopoietic stem cells
- Author
-
Louis M. Pelus and Hal E. Broxmeyer
- Subjects
business.industry ,Lethal dose ,Hematopoietic stem cell ,Context (language use) ,Cell Biology ,Hematology ,Transplantation ,Haematopoiesis ,surgical procedures, operative ,medicine.anatomical_structure ,Cord blood ,Immunology ,medicine ,Cancer research ,Molecular Medicine ,Bone marrow ,Stem cell ,business ,Molecular Biology - Abstract
Enhancing the engraftment of hematopoietic stem cells (HSC) is especially important when times to engraftment are prolonged due either to limiting numbers of HSC in the donor graft or to intrinsic slower engrafting time of the tissue sources of HSC. Both inhibition of dipeptidylpeptidase (DPP) 4/CD26 and treatment of cells with 16,16 dimethyl prostaglandin E2 (dmPGE2) have been shown to enhance hematopoietic stem cell engraftment in murine transplantation models and have been evaluated in clinical settings for their influence on engraftment of cord blood cells, a tissue source of HSC known to manifest an extended time to engraftment of donor cells compared to that of bone marrow (BM) and mobilized peripheral blood for hematopoietic cell transplantation (HCT). Herein, we present new experimental data, using a CD45(+) head-to-head congenic model of donor mouse BM cells for engraftment of lethally irradiated mice, demonstrating that similar levels of enhanced engraftment are detected by pulsing donor BM cells with diprotin A, a DPP4 inhibitor, or with dmPGE2 prior to infusion, or by pretreating recipient mice with sitagliptin, also a DPP4 inhibitor, by oral gavage. Moreover, the combined effects of pretreating the donor BM cells with dmPGE2 in context of pretreating the recipient mice with sitagliptin after the administration of a lethal dose of radiation resulted in significantly enhanced competitively repopulating HCT compared to either treatment alone. This information is highly relevant to the goal of enhancing engraftment in human clinical HCT.
- Published
- 2014
- Full Text
- View/download PDF
34. Spotlight on Glycolysis: A New Target for Cord Blood Expansion
- Author
-
Andrea M. Patterson and Louis M. Pelus
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Peroxisome proliferator-activated receptor ,Hematopoietic stem cell transplantation ,Biology ,Umbilical cord ,Article ,03 medical and health sciences ,fluids and secretions ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Glycolysis ,chemistry.chemical_classification ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Fetal Blood ,Hematopoietic Stem Cells ,PPAR gamma ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Cord blood ,embryonic structures ,Cancer research ,Molecular Medicine ,Stem cell ,Antagonism - Abstract
Hematopoietic stem cells (HSCs) quiescently reside in bone marrow niches and have the capacity to self-renew or differentiate to form all blood cells throughout the lifespan of an animal1–3. Allogeneic HSC transplantation is a life-saving treatment for malignant and non-malignant disorders4,5. HSCs isolated from umbilical cord blood (CB) are used for hematopoietic cell transplantation (HCT)6–11, but due to limited numbers of HSCs in single units of umbilical CB, a number of methods have been proposed for ex vivo expansion of human HSCs7,8,12. We show here that antagonism of the nuclear hormone receptor PPARγ promotes ex vivo expansion of phenotypically and functionally-defined subsets of human CB HSCs and hematopoietic progenitor cells (HSPCs). PPARγ antagonism in CB HSPCs strongly downregulated expression of several differentiation associated genes, as well as fructose 1, 6-bisphosphatase (FBP1), a negative regulator of glycolysis, and enhanced glycolysis without compromising mitochondrial metabolism. The expansion of CB HSPCs by PPARγ antagonism was completely suppressed by removal of glucose or inhibition of glycolysis. Moreover, knockdown of FBP1 expression promoted glycolysis and ex vivo expansion of long-term repopulating CB HSPCs, whereas overexpression of FBP1 suppressed the expansion of CB HSPCs induced by PPARγ antagonism. Our study suggests the possibility for a new and simple means for metabolic reprogramming of CB HSPCs to improve the efficacy of HCT.
- Published
- 2018
- Full Text
- View/download PDF
35. Concise Review: Sowing the Seeds of a Fruitful Harvest: Hematopoietic Stem Cell Mobilization
- Author
-
Jennifer M. Speth, Louis M. Pelus, and Jonathan Hoggatt
- Subjects
medicine.medical_treatment ,Hematopoietic stem cell ,Cell Biology ,Hematopoietic stem cell transplantation ,Biology ,Bioinformatics ,CXCR4 ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,medicine ,Molecular Medicine ,Autologous transplantation ,Progenitor cell ,Hematopoietic Stem Cell Mobilization ,Developmental Biology - Abstract
Hematopoietic stem cell transplantation is the only curative option for a number of malignant and nonmalignant diseases. As the use of hematopoietic transplant has expanded, so too has the source of stem and progenitor cells. The predominate source of stem and progenitors today, particularly in settings of autologous transplantation, is mobilized peripheral blood. This review will highlight the historical advances which led to the widespread use of peripheral blood stem cells for transplantation, with a look toward future enhancements to mobilization strategies. Stem Cells 2013;31:2599–2606
- Published
- 2013
- Full Text
- View/download PDF
36. Prostaglandin E2 enhances long-term repopulation but does not permanently alter inherent stem cell competitiveness
- Author
-
Pratibha Singh, Louis M. Pelus, Jonathan Hoggatt, and Khalid S. Mohammad
- Subjects
Hematopoiesis and Stem Cells ,medicine.medical_treatment ,Immunology ,Cell Count ,Hematopoietic stem cell transplantation ,Biology ,Biochemistry ,Dinoprostone ,Mice ,medicine ,Animals ,Cell Lineage ,Prostaglandin E2 ,Cells, Cultured ,Cell Proliferation ,Cell growth ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,hemic and immune systems ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,surgical procedures, operative ,medicine.anatomical_structure ,Cancer research ,lipids (amino acids, peptides, and proteins) ,Stem cell ,Homing (hematopoietic) ,medicine.drug - Abstract
Hematopoietic stem cell (HSC) transplantation is a lifesaving therapy for malignant and nonmalignant hematologic diseases and metabolic disorders. Although successful, hematopoietic transplantation can be hindered by inadequate stem cell number or poor engrafting efficiency. To overcome these deficits, we and others have previously reported the HSC-enhancing ability of a short-term exposure of prostaglandin E2 (PGE2); this strategy has now progressed to phase 1 clinical trials in double cord blood transplantation. To further analyze the short- and long-term effects of HSC exposure to PGE2, we followed the repopulation kinetics of PGE2-treated hematopoietic grafts through 5 serial transplantations and compared inherent long-term competitiveness in a HSC head-to-head secondary transplantation model. Treatment with PGE2 did not result in a long-term increase in HSC competitiveness, lineage bias, or enhanced proliferative potential, demonstrating that pulse exposure to PGE2 results in transient increases in HSC homing and engraftment potential.
- Published
- 2013
- Full Text
- View/download PDF
37. New G-CSF agonists for neutropenia therapy
- Author
-
Jonathan Hoggatt and Louis M. Pelus
- Subjects
Pharmacology ,medicine.medical_specialty ,Myelosuppressive Chemotherapy ,Neutropenia ,business.industry ,Cost effectiveness ,General Medicine ,Filgrastim ,Granulocyte ,medicine.disease ,Haematopoiesis ,medicine.anatomical_structure ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Intensive care medicine ,business ,Biosimilar Pharmaceuticals ,Pegfilgrastim ,Febrile neutropenia ,medicine.drug - Abstract
Granulocyte colony-stimulating factor (G-CSF; filgrastim) and its pegylated form (pegfilgrastim) are widely used to treat neutropenia associated with myelosuppressive chemotherapy and bone marrow transplantation, AIDS-associated or drug-induced neutropenia, and neutropenic diseases. G-CSF facilitates restoration of neutrophil counts, decreases incidence of infection/febrile neutropenia and reduces resource utilization. G-CSF is also widely used to mobilize peripheral blood stem cells for hematopoietic transplant.We review the therapeutic use, cost effectiveness and disease impact of G-CSF for neutropenia, development of G-CSF biosimilars and current next-generation discovery efforts.G-CSF has impacted the treatment and survival of patients with congenital neutropenias. For chemotherapy-associated neutropenia, cost effectiveness and impact on survival are still unclear. G-CSFs are expensive and require systemic administration. Market entry of new biosimilars, some with enhanced half-life profiles, will probably reduce cost and increase cost effectiveness. There is no evidence that marketed or late development biosimilars display effectiveness superior to current G-CSFs. Second-generation compounds that mimic the activity of G-CSF at its receptor, induce endogenous ligand(s) or offer adjunct activity have been reported and represent attractive G-CSF alternatives, but are in preclinical stages. A significant therapeutic advance will require reduced depth and duration of neutropenia compared to current G-CSFs.
- Published
- 2013
- Full Text
- View/download PDF
38. Small molecule inhibition of CBP/catenin interactions eliminates drug resistant clones in acute lymphoblastic leukemia
- Author
-
Eugene Park, Michael McMillan, Sandra Huantes, Srividya Swaminathan, Jennifer Pham, Michael Kahn, Yao-Te Hsieh, Khatija Naing, Edward M. Conway, Lars Klemm, Louis M. Pelus, John D. Crispino, Markus Müschen, Yi Zhao, Cu Nguyen, Charles G. Mullighan, Eun Ji Gang, and Yong-Mi Kim
- Subjects
Cancer Research ,Survivin ,Mice, SCID ,Acute lymphoblastic leukemia ,Dexamethasone ,Inhibitor of Apoptosis Proteins ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Wnt Signaling Pathway ,beta Catenin ,0303 health sciences ,biology ,ICG-001 ,Wnt signaling pathway ,Myeloid leukemia ,small molecule inhibitor ,Drug Synergism ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,3. Good health ,Leukemia ,Vincristine ,030220 oncology & carcinogenesis ,Beta-catenin ,Cell Survival ,Sialoglycoproteins ,Down-Regulation ,p300 ,Antineoplastic Agents ,Pyrimidinones ,CBP ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Coactivator ,Genetics ,medicine ,Animals ,Asparaginase ,Humans ,EP300 ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,drug resistance ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Xenograft Model Antitumor Assays ,Peptide Fragments ,Drug Resistance, Neoplasm ,Catenin ,Mutation ,biology.protein ,Cancer research ,Chromatin immunoprecipitation - Abstract
Drug resistance in acute lymphoblastic leukemia (ALL) remains a major problem warranting new treatment strategies. Wnt/catenin signaling is critical for the self-renewal of normal hematopoietic progenitor cells. Deregulated Wnt signaling is evident in chronic and acute myeloid leukemia; however, little is known about ALL. Differential interaction of catenin with either the Kat3 coactivator CREBBP (CREB-binding protein (CBP)) or the highly homologous EP300 (p300) is critical to determine divergent cellular responses and provides a rationale for the regulation of both proliferation and differentiation by the Wnt signaling pathway. Usage of the coactivator CBP by catenin leads to transcriptional activation of cassettes of genes that are involved in maintenance of progenitor cell self-renewal. However, the use of the coactivator p300 leads to activation of genes involved in the initiation of differentiation. ICG-001 is a novel small-molecule modulator of Wnt/catenin signaling, which specifically binds to the N-terminus of CBP and not p300, within amino acids 1–110, thereby disrupting the interaction between CBP and catenin. Here, we report that selective disruption of the CBP/β- and γ-catenin interactions using ICG-001 leads to differentiation of pre-B ALL cells and loss of self-renewal capacity. Survivin, an inhibitor-of-apoptosis protein, was also downregulated in primary ALL after treatment with ICG-001. Using chromatin immunoprecipitation assay, we demonstrate occupancy of the survivin promoter by CBP that is decreased by ICG-001 in primary ALL. CBP mutations have been recently identified in a significant percentage of ALL patients, however, almost all of the identified mutations reported occur C-terminal to the binding site for ICG-001. Importantly, ICG-001, regardless of CBP mutational status and chromosomal aberration, leads to eradication of drug-resistant primary leukemia in combination with conventional therapy in vitro and significantly prolongs the survival of NOD/SCID mice engrafted with primary ALL. Therefore, specifically inhibiting CBP/catenin transcription represents a novel approach to overcome relapse in ALL.
- Published
- 2013
39. Survivin Is Required for Mouse and Human Bone Marrow Mesenchymal Stromal Cell Function
- Author
-
Liqiong Liu, Seiji Fukuda, Brahmananda R. Chitteti, Pratibha Singh, and Louis M. Pelus
- Subjects
0301 basic medicine ,Stromal cell ,Platelet-derived growth factor ,Survivin ,Basic fibroblast growth factor ,Biology ,Inhibitor of Apoptosis Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Cell Movement ,Animals ,Humans ,Cell Proliferation ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Mice, Inbred C57BL ,Repressor Proteins ,030104 developmental biology ,chemistry ,Immunology ,Cancer research ,Molecular Medicine ,Stem cell ,Wound healing ,Developmental Biology ,Adult stem cell - Abstract
Although mesenchymal stromal cells (MSCs) have significant potential in cell-based therapies, little is known about the factors that regulate their functions. While exploring regulatory molecules potentially involved in MSC activities, we found that the endogenous multifunctional factor Survivin is essential for MSC survival, expansion, lineage commitment, and migration. Pharmacological or genetic blockade of Survivin expression in mouse and human bone marrow MSC enhances caspase 3 and 7 expression and reduces proliferation resulting in fewer MSC and clonogenic colony-forming unit-fibroblasts (CFU-F), whereas ectopic Survivin overexpression in MSC results in their expansion. Survivin is also required for the MSC proliferative responses to basic fibroblast growth factor and platelet derived growth factor. In a wound healing model, Survivin inhibition results in suppression of MSC migration to the wound site. In addition, loss of Survivin in MSCs compromises their hematopoiesis-supporting capacity. These results demonstrate that Survivin is a key regulator of mouse and human MSC function, and suggest that targeted modulation of Survivin in MSCs may have clinical utility to enhance MSC recovery and activity following insult or stress.
- Published
- 2016
40. Dipeptidylpeptidase 4 negatively regulates colony-stimulating factor activity and stress hematopoiesis
- Author
-
Jonathan Hoggatt, Giao Hangoc, Steven Messina-Graham, Sara L. Rohrabaugh, Sherif S. Farag, Brahmananda R. Chitteti, Louis M. Pelus, Heather O'Leary, Xuan Ou, Jennifer M. Speth, Hal E. Broxmeyer, Timothy B. Campbell, Scott Cooper, Charlie Mantel, and Edward F. Srour
- Subjects
medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Dipeptidyl Peptidase 4 ,medicine.medical_treatment ,Biology ,Mass Spectrometry ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Immunophenotyping ,Mice ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,Progenitor cell ,DNA Primers ,Mice, Knockout ,Radiotherapy ,Granulocyte-Macrophage Colony-Stimulating Factor ,Hematopoietic stem cell ,General Medicine ,Colony-stimulating factor ,Chemokine CXCL12 ,Hematopoiesis ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,Cytokine ,Endocrinology ,medicine.anatomical_structure ,Cancer research ,Ex vivo ,Signal Transduction - Abstract
Enhancement of hematopoietic recovery after radiation, chemotherapy, or hematopoietic stem cell (HSC) transplantation is clinically relevant. Dipeptidylpeptidase (DPP4) cleaves a wide variety of substrates, including the chemokine stromal cell-derived factor-1 (SDF-1). In the course of experiments showing that inhibition of DPP4 enhances SDF-1-mediated progenitor cell survival, ex vivo cytokine expansion and replating frequency, we unexpectedly found that DPP4 has a more general role in regulating colony-stimulating factor (CSF) activity. DPP4 cleaved within the N-termini of the CSFs granulocyte-macrophage (GM)-CSF, G-CSF, interleukin-3 (IL-3) and erythropoietin and decreased their activity. Dpp4 knockout or DPP4 inhibition enhanced CSF activities both in vitro and in vivo. The reduced activity of DPP4-truncated versus full-length human GM-CSF was mechanistically linked to effects on receptor-binding affinity, induction of GM-CSF receptor oligomerization and signaling capacity. Hematopoiesis in mice after radiation or chemotherapy was enhanced in Dpp4(-/-) mice or mice receiving an orally active DPP4 inhibitor. DPP4 inhibition enhanced engraftment in mice without compromising HSC function, suggesting the potential clinical utility of this approach.
- Published
- 2012
- Full Text
- View/download PDF
41. Blockade of prostaglandin E-2 signaling through EP1 and EP3 receptors attenuates Flt3L-dependent dendritic cell development from hematopoietic progenitor cells
- Author
-
Pratibha Singh, Richard M. Breyer, Seiji Fukuda, Peirong Hu, Jennifer M. Speth, Louis M. Pelus, and Jonathan Hoggatt
- Subjects
STAT3 Transcription Factor ,Hematopoiesis and Stem Cells ,Cellular differentiation ,Survivin ,Immunology ,Biology ,Biochemistry ,Dinoprostone ,Inhibitor of Apoptosis Proteins ,Mice ,Hormone Antagonists ,Animals ,Humans ,Progenitor cell ,Receptor ,Cells, Cultured ,Progenitor ,Mice, Knockout ,Infant, Newborn ,Membrane Proteins ,Cell Differentiation ,Cell Biology ,Hematology ,Dendritic cell ,Dendritic Cells ,Hematopoietic Stem Cells ,Receptors, Prostaglandin E, EP1 Subtype ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Receptors, Prostaglandin E, EP3 Subtype ,lipids (amino acids, peptides, and proteins) ,Signal transduction ,Signal Transduction - Abstract
Dendritic cell (DC) homeostasis, like all mature blood cells, is maintained via hierarchal generation from hematopoietic precursors; however, little is known about the regulatory mechanisms governing DC generation. Here, we show that prostaglandin E2 (PGE2) is required for optimal Flt3 ligand–mediated DC development and regulates expression of the Flt3 receptor on DC-committed progenitor cells. Inhibition of PGE2 biosynthesis reduces Flt3-mediated activation of STAT3 and expression of the antiapoptotic protein survivin, resulting in increased apoptosis of DC-committed progenitor cells. Reduced DC development caused by diminished PGE2 signaling is reversed by overexpression of Flt3 or survivin in DC progenitors and conversely is mimicked by STAT3 inhibition. PGE2 regulation of DC generation is specifically mediated through the EP1 and EP3 G protein PGE2 receptors. These studies define a novel DC progenitor regulatory pathway in which PGE2 signaling through EP1/EP3 receptors regulates Flt3 expression and downstream STAT3 activation and survivin expression, required for optimal DC progenitor survival and DC development in vivo.
- Published
- 2012
42. Pleiotropic effects of prostaglandin E2 in hematopoiesis; prostaglandin E2 and other eicosanoids regulate hematopoietic stem and progenitor cell function
- Author
-
Louis M. Pelus and Jonathan Hoggatt
- Subjects
Receptors, CXCR4 ,Physiology ,medicine.medical_treatment ,Apoptosis ,Stem cell factor ,Hematopoietic stem cell transplantation ,Biology ,Biochemistry ,Article ,Dinoprostone ,Mice ,Cell Movement ,medicine ,Animals ,Humans ,Yin-Yang ,Cell Lineage ,Progenitor cell ,Pharmacology ,Cannabinoids ,Cell Cycle ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Cell Biology ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Hematopoiesis ,Cell biology ,Endothelial stem cell ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Gene Expression Regulation ,lipids (amino acids, peptides, and proteins) ,Stem cell ,Signal Transduction - Abstract
Eicosanoids have been implicated in the physiological regulation of hematopoiesis with pleiotropic effects on hematopoietic stem cells and various classes of lineage restricted progenitor cells. Herein we review the effects of eicosanoids on hematopoiesis, focusing on new findings implicating prostaglandin E(2) in enhancing hematopoietic stem cell engraftment by enhancing stem cell homing, survival and self-renewal. We also describe a role for cannabinoids in hematopoiesis. Lastly, we discuss the yin and yang of various eicosanoids in modulating hematopoietic stem and progenitor cell functions and summarize potential strategies to take advantage of these effects for therapeutic benefit for hematopoietic stem cell transplantation.
- Published
- 2011
- Full Text
- View/download PDF
43. Targeting survivin overcomes drug resistance in acute lymphoblastic leukemia
- Author
-
Wolf-Karsten Hofmann, Eun Suk Kang, Sandra Huantes, Edward M. Conway, Lars Klemm, Yong-Mi Kim, Eugene Park, Nora Heisterkamp, Ganesan Keerthivasan, Paul Schaefer, Mignon L. Loh, Hong Hoe Koo, Michael Kahn, Sanna Chae, John D. Crispino, Yao Te Hsieh, Louis M. Pelus, Markus Müschen, and Eun Ji Gang
- Subjects
Neoplasm, Residual ,Survivin ,Immunology ,Oligonucleotides ,Gene Expression ,Drug resistance ,Biology ,Inhibitor of apoptosis ,Biochemistry ,Inhibitor of Apoptosis Proteins ,Small hairpin RNA ,Mice ,Mice, Inbred NOD ,Acute lymphocytic leukemia ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Tumor Stem Cell Assay ,Mice, Knockout ,Lymphoid Neoplasia ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Chemotherapy regimen ,Minimal residual disease ,Repressor Proteins ,Leukemia ,Drug Resistance, Neoplasm ,Gene Targeting ,Cancer research - Abstract
Relapse of drug-resistant acute lymphoblastic leukemia (ALL) has been associated with increased expression of survivin/BIRC5, an inhibitor of apoptosis protein, suggesting a survival advantage for ALL cells. In the present study, we report that inhibition of survivin in patient-derived ALL can eradicate leukemia. Targeting survivin with shRNA in combination with chemotherapy resulted in no detectable minimal residual disease in a xenograft model of primary ALL. Similarly, pharmacologic knock-down of survivin using EZN-3042, a novel locked nucleic acid antisense oligonucleotide, in combination with chemotherapy eliminated drug-resistant ALL cells. These findings show the importance of survivin expression in drug resistance and demonstrate that survivin inhibition may represent a powerful approach to overcoming drug resistance and preventing relapse in patients with ALL.
- Published
- 2011
- Full Text
- View/download PDF
44. Many mechanisms mediating mobilization: an alliterative review
- Author
-
Jonathan Hoggatt and Louis M. Pelus
- Subjects
medicine.medical_specialty ,Hematology ,Niche ,Hematopoietic stem cell ,Biology ,Hematopoietic Stem Cells ,Article ,Hematopoietic Stem Cell Mobilization ,Granulocyte colony-stimulating factor ,Cell biology ,Mice ,Haematopoiesis ,medicine.anatomical_structure ,Cell Movement ,Internal medicine ,Immunology ,medicine ,Animals ,Humans ,Bone marrow ,Stem Cell Niche ,Stem cell - Abstract
Blood cell production is maintained by hematopoietic stem cells (HSCs) that reside in specialized niches within bone marrow. Treatment with granulocyte-colony stimulating factor (G-CSF) causes HSC egress from bone marrow niches and trafficking to the peripheral blood, a process termed 'mobilization'. Although the mobilization phenomenon has been known for some time and is utilized clinically to acquire HSC for transplant, the mechanisms mediating HSC release are not completely understood. We discuss recent advances and controversies in defining the mechanisms responsible for G-CSF-induced mobilization.New reports define a role for resident monocytes/macrophages in maintaining niche cells, which is diminished after G-CSF treatment, suggesting a new mechanism for mobilization. Although osteoblasts have been reported to be a primary component of the HSC niche, new results suggest a unique niche composed of innervated mesenchymal stem cells. Modulating bioactive lipid signaling also facilitates mobilization, and may define a future therapeutic strategy.Hematopoietic mobilization by G-CSF is primarily mediated by alterations to the bone marrow niche by both direct and indirect mechanisms, rather than directly altering HSC function. Further understanding of the processes mediating mobilization will advance our understanding on the cellular and molecular components of the HSC niche.
- Published
- 2011
- Full Text
- View/download PDF
45. Pulse exposure of haematopoietic grafts to prostaglandin E2in vitro facilitates engraftment and recovery
- Author
-
Louis M. Pelus, Pratibha Singh, and Jonathan Hoggatt
- Subjects
medicine.medical_treatment ,Prostaglandin ,Cell Biology ,General Medicine ,Biology ,Transplantation ,chemistry.chemical_compound ,Haematopoiesis ,chemistry ,In vivo ,Immunology ,Survivin ,Cancer research ,medicine ,lipids (amino acids, peptides, and proteins) ,Stem cell ,Ex vivo ,Prostaglandin E - Abstract
Objectives The aim of this study was to evaluate the effects of prostaglandin E(2) (PGE(2) ) on haematopoietic stem cell (HSC) function and determine its mechanism of action. Materials and methods HSC were exposed to PGE(2) for 2 h and effects on their homing, engraftment and self-renewal evaluated in vivo. Effects of PGE(2) on HSC cell cycle, CXCR4 expression and migration to SDF-1α were analysed in vitro. Apoptosis was evaluated by examination of survivin expression and active caspase-3 levels. Results Equivalent haematopoietic reconstitution was demonstrated using 4-fold fewer PGE(2) -treated cells compared to controls. Multilineage reconstitution was stable on secondary transplantation, indicating that PGE(2) affects long-term repopulating HSC (LT-HSC) and that enhanced chimaerism of PGE(2) -pulsed cells results from their initial treatment. PGE(2) increased CXCR4 expression on mouse and human HSC, increased their migration to SDF-1αin vitro and enhanced in vivo marrow homing 2-fold, which was blocked by a CXCR4 receptor antagonist. PGE(2) pulse exposure reduced apoptosis of mouse and human HSC, with increase in endogenous caspase inhibitor survivin, and concomitant decrease in active caspase-3. Two-fold more HSC entered the cell cycle and proliferated within 24 h after PGE(2) pulse exposure. Conclusions These studies demonstrate that short-term PGE(2) exposure enhances HSC function and supports the concept of utility of PGE(2) as an ex vivo strategy to improve function of haematopoietic grafts, particularly those where HSC numbers are limited.
- Published
- 2011
- Full Text
- View/download PDF
46. Increased mobilization and yield of stem cells using plerixafor in combination with granulocyte-colony stimulating factor for the treatment of non-Hodgkin’s lymphoma and multiple myeloma
- Author
-
Louis M. Pelus and Sherif S. Farag
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Medicine (miscellaneous) ,lymphoma ,Review ,CXCR4 ,stem cells ,Internal medicine ,medicine ,Progenitor cell ,mobilization ,Multiple myeloma ,QH573-671 ,business.industry ,Plerixafor ,plerixafor ,Cell Biology ,medicine.disease ,Granulocyte colony-stimulating factor ,Non-Hodgkin's lymphoma ,medicine.anatomical_structure ,myeloma ,Immunology ,Stem cell ,business ,Cytology ,medicine.drug - Abstract
Louis M Pelus1, Sherif S Farag21Department of Microbiology and Immunology, 2Division of Hematology and Oncology, Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IndianaAbstract: Multiple myeloma and non-Hodgkin’s lymphoma remain the most common indications for high-dose chemotherapy and autologous peripheral blood stem cell rescue. While a CD34+ cell dose of 1 × 106/kg is considered the minimum required for engraftment, higher CD34+ doses correlate with improved outcome. Numerous studies, however, support targeting a minimum CD34+ cell dose of 2.0 × 106/kg, and an “optimal” dose of 4 to 6 × 106/kg for a single transplant. Unfortunately, up to 40% of patients fail to mobilize an optimal CD34+ cell dose using myeloid growth factors alone. Plerixafor is a novel reversible inhibitor of CXCR4 that significantly increases the mobilization and collection of higher numbers of hematopoietic progenitor cells. Two randomized multi-center clinical trials in patients with non-Hodgkin’s lymphoma and multiple myeloma have demonstrated that the addition of plerixafor to granulocyte-colony stimulating factor increases the mobilization and yield of CD34+ cells in fewer apheresis days, which results in durable engraftment. This review summarizes the pharmacology and evidence for the clinical efficacy of plerixafor in mobilizing hematopoietic stem and progenitor cells, and discusses potential ways to utilize plerixafor in a cost-effective manner in patients with these diseases.Keywords: plerixafor, mobilization, stem cells, lymphoma, myeloma
- Published
- 2011
47. Engraftment and Reconstitution of Hematopoiesis Is Dependent on Endothelial Cell Expressed Prostaglandin EP4 Receptor Signaling Mediated Regeneration of Vascular Niche
- Author
-
Louis M. Pelus, Hongge Li, and Pratibha Singh
- Subjects
Stromal cell ,Leptin receptor ,Immunology ,Mesenchymal stem cell ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Endothelial stem cell ,Transplantation ,Haematopoiesis ,Progenitor cell ,Stem cell - Abstract
Defining the cellular and molecular components of the bone marrow (BM) niche that regulate the homeostasis and regeneration of hematopoietic stem and progenitor cells (HSPCs) can facilitate targeted interventions to improve hematopoietic recovery after injury and enhance stem cell engraftment after transplantation. We have recently shown that BM niche prostaglandin E2 (PGE2) signaling via the EP4 receptor is crucial for hematopoietic reconstitution, as transplantation of wild-type donor cells into EP4 receptor deficient recipient mice shows reduced stem cell engraftment. However, the specific contribution of EP4 signaling on different niche constituents in hematopoietic regeneration is unknown. To investigate which niche constituent/stromal population EP4 signaling functionally contributes to hematopoietic regeneration, we utilized genetic mouse models, in which EP4 could be deleted in specific stromal cell types. We conditionally deleted EP4 expression in nestin expressing mesenchymal stem cell (MSC), leptin receptor expressing mesenchymal stromal cell or tie 2 expressing endothelial cell (EC) invivo using a cre-recombinase approach. At steady-state, selective deletion of EP4 from nestin+ MSC (nestin-cre EP4flox/flox mice), leptin receptor+stromal cell (leptin R-cre EP4flox/flox mice) or tie 2+EC (tie 2-cre EP4flox/flox mice) showed no change in BM MSC (CD45-Ter119-CD31-CD51+PDGFR+), leptin receptor+stromal cells (CD45-Ter119-leptin R+) and EC (CD45-ter119-CD31+VE-cadherin+) number and survival. We next determined whether loss of EP4 expression/signaling in stromal cell specific genetic knockout mouse models affects BM niche constituents after exposure to ionization radiation. Interestingly, loss of EC but not nestin/leptin EP4 expression severely disrupted the BM vasculature and reduced EC and MSC survival at 24 hours post total body lethal (TBI) irradiation, compared to wild-type irradiated mice, and was associated with reduced expression of Survivin and increased expression of Bax, anti-apoptotic and pro-apoptotic factors respectively. In addition, niche EC and MSC regeneration at 2-month post irradiation was substantially reduced in tie 2-cre EP4flox/flox mice compared to wild-type mice. Nestin-cre EP4flox/flox mice and leptin R-cre EP4flox/flox niche restoration was equivalent to wild-type mice. To determine whether loss of EP4 receptor in different niche cells differentially regulates hematopoietic regeneration, we transplanted wild-type donor cells into nestin-cre EP4flox/flox or tie 2-cre EP4flox/flox recipient mice. Chimeric mice made with EC-specific EP4 deficient stroma showed reduced HSPC engraftment compared to chimeric mice made with wild-type stroma. In contrast, stem cell engraftment in chimeric mice made with MSC-specific EP4 deficient stroma was not reduced. In addition, in vivo administration of PGE2 into wild-type recipient mice at six-hour post irradiation enhanced donor cell engraftment. However, PGE2 treatment failed to enhance donor cell engraftment into tie 2-cre EP4flox/flox recipient mice. These data suggest that EC EP4 expression is important for niche restoration and hematopoietic regeneration after transplantation. To identify the mechanism involved in EP4 signaling mediated niche restoration, we measured angiocrine factors in the BM niche of lethally irradiated wild-type and tie 2-cre EP4flox/flox mice. Interestingly, we found that vascular endothelial growth factor (VEGF) expression was substantially reduced in the BM of tie 2-cre EP4flox/flox mice compared to wild-type mice. To investigate whether EP4 signaling regulates niche restoration by stimulating VEGF expression/production, we treated lethally irradiated tie 2-cre EP4flox/flox mice with recombinant VEGF (20 ng/mouse/day) for 3 days. In vivo administration of recombinant VEGF substantially enhanced niche restoration in tie 2-cre EP4flox/floxmice compared to control tie 2-cre EP4flox/flox. In conclusion, our study suggests that EC specific EP4 receptor signaling-mediated increased expression of VEGF promotes vascular niche restoration after myelosuppression and supports hematopoietic regeneration. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
48. Eicosanoid regulation of hematopoiesis and hematopoietic stem and progenitor trafficking
- Author
-
Jonathan Hoggatt and Louis M. Pelus
- Subjects
Leukotrienes ,Cancer Research ,Receptor expression ,CD34 ,Biology ,CXCR4 ,Dinoprostone ,Article ,Cell Movement ,medicine ,Animals ,Humans ,Yin-Yang ,Cannabinoids ,Hematopoietic stem cell ,hemic and immune systems ,Hematology ,Hematopoietic Stem Cells ,Hematopoiesis ,Cell biology ,Transplantation ,medicine.anatomical_structure ,Oncology ,Immunology ,Eicosanoids ,lipids (amino acids, peptides, and proteins) ,Bone marrow ,Stem cell ,Homing (hematopoietic) - Abstract
Hematopoietic stem cell (HSC) transplantation is a potentially curative treatment for numerous hematological malignancies. The transplant procedure as performed today takes advantage of HSC trafficking; either egress of HSC from the bone marrow to the peripheral blood, that is, mobilization, for acquisition of the hematopoietic graft, and/or trafficking of HSC from the peripheral blood to bone marrow niches in the recipient patient, that is HSC homing. Numerous studies, many of which are reviewed herein, have defined hematopoietic regulatory mechanisms mediated by the 20-carbon lipid family of eicosanoids, and recent evidence strongly supports a role for eicosanoids in regulation of hematopoietic trafficking, adding a new role whereby eicosanoids regulate hematopoiesis. Short-term exposure of HSC to the eicosanoid prostaglandin E(2) increases CXCR4 receptor expression, migration and in vivo homing of HSC. In contrast, cannabinoids reduce hematopoietic progenitor cell (HPC) CXCR4 expression and induce HPC mobilization when administered in vivo. Leukotrienes have been shown to alter CD34(+) cell adhesion, migration and regulate HSC proliferation, suggesting that eicosanoids have both opposing and complimentary roles in the regulation of hematopoiesis. As numerous FDA approved compounds regulate eicosanoid signaling or biosynthesis, the utility of eicosanoid-based therapeutic strategies to improve hematopoietic transplantation can be rapidly evaluated.
- Published
- 2010
- Full Text
- View/download PDF
49. Hematopoietic Agents
- Author
-
Louis M. Pelus, Jonathan Hoggatt, Pratibha Singh, and Janardhan Sampath
- Published
- 2010
- Full Text
- View/download PDF
50. The chemokine GROβ mobilizes early hematopoietic stem cells characterized by enhanced homing and engraftment
- Author
-
Andrew G. King, Louis M. Pelus, Huimin Bian, and Seiji Fukuda
- Subjects
Receptors, CXCR4 ,Stromal cell ,Chemokine CXCL1 ,Dipeptidyl Peptidase 4 ,Immunology ,Vascular Cell Adhesion Molecule-1 ,Antigens, CD34 ,Biology ,Biochemistry ,CXCR4 ,Mice ,Cell Movement ,Granulocyte Colony-Stimulating Factor ,Cell Adhesion ,medicine ,Animals ,Antigens, Ly ,Transplantation, Homologous ,Hematopoietic Stem Cell Mobilization ,Mice, Knockout ,Mice, Inbred BALB C ,Peripheral Blood Stem Cell Transplantation ,Graft Survival ,Endothelial Cells ,Membrane Proteins ,Recovery of Function ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Hematopoiesis ,Granulocyte colony-stimulating factor ,Proto-Oncogene Proteins c-kit ,Haematopoiesis ,medicine.anatomical_structure ,Gene Expression Regulation ,Cancer research ,Female ,Bone marrow ,Stem cell ,Chemokines, CXC ,Homing (hematopoietic) - Abstract
Mobilized peripheral blood hematopoietic stem cells (PBSCs) demonstrate accelerated engraftment compared with bone marrow; however, mechanisms responsible for enhanced engraftment remain unknown. PBSCs mobilized by GRObeta (GRObeta(Delta4)/CXCL2(Delta4)) or the combination of GRObeta(Delta4) plus granulocyte colony-stimulating factor (G-CSF) restore neutrophil and platelet recovery faster than G-CSF-mobilized PBSCs. To determine mechanisms responsible for faster hematopoietic recovery, we characterized immunophenotype and function of the GRObeta-mobilized grafts. PBSCs mobilized by GRObeta(Delta4) alone or with G-CSF contained significantly more Sca-1(+)-c-kit(+)-lineage(-) (SKL) cells and more primitive CD34(-)-SKL cells compared with cells mobilized by G-CSF and demonstrated superior competitive long-term repopulation activity, which continued to increase in secondary and tertiary recipients. GRObeta(Delta4)-mobilized SKL cells adhered better to VCAM-1(+) endothelial cells compared with G-CSF-mobilized cells. GRObeta(Delta4)-mobilized PBSCs did not migrate well to the chemokine stromal derived factor (SDF)-1alpha in vitro that was associated with higher CD26 expression. However, GRObeta(Delta4)-mobilized SKL and c-Kit(+) lineage(-) (KL) cells homed more efficiently to marrow in vivo, which was not affected by selective CXCR4 and CD26 antagonists. These data suggest that GRObeta(Delta4)-mobilized PBSCs are superior in reconstituting long-term hematopoiesis, which results from differential mobilization of early stem cells with enhanced homing and long-term repopulating capacity. In addition, homing and engraftment of GRObeta(Delta4)-mobilized cells is less dependent on the SDF-1alpha/CXCR4 axis.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.