29 results on '"Lisa M. Niswander"'
Search Results
2. Potent preclinical activity of FLT3-directed chimeric antigen receptor T-cell immunotherapy against FLT3- mutant acute myeloid leukemia and KMT2A-rearranged acute lymphoblastic leukemia
- Author
-
Lisa M. Niswander, Zachary T. Graff, Christopher D. Chien, John A. Chukinas, Christina A. Meadows, Lillie C. Leach, Joseph P. Loftus, M. Eric Kohler, Sarah K. Tasian, and Terry J. Fry
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapies targeting CD19 or CD22 induce remissions in the majority of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL), although relapse due to target antigen loss or downregulation has emerged as a major clinical dilemma. Accordingly, great interest exists in developing CAR T cells directed against alternative leukemia cell surface antigens that may help to overcome immunotherapeutic resistance. The fms-like tyrosine kinase 3 receptor (FLT3) is constitutively activated via FLT3 mutation in acute myeloid leukemia (AML) or wild-type FLT3 overexpression in KMT2A (lysine-specific methyltransferase 2A)-rearranged ALL, which are associated with poor clinical outcomes in children and adults. We developed monovalent FLT3-targeted CAR T cells (FLT3CART) and bispecific CD19xFLT3CART and assessed their anti-leukemia activity in preclinical models of FLT3-mutant AML and KMT2A-rearranged infant ALL. We report robust in vitro FLT3CART-induced cytokine production and cytotoxicity against AML and ALL cell lines with minimal cross-reactivity against normal hematopoietic and non-hematopoietic tissues. We also observed potent in vivo inhibition of leukemia proliferation in xenograft models of both FLT3-mutant AML and KMT2A-rearranged ALL, including a post-tisagenlecleucel ALL-to-AML lineage switch patient-derived xenograft model pairing. We further demonstrate significant in vitro and in vivo activity of bispecific CD19xFLT3CART against KMT2Arearranged ALL and posit that this additional approach might also diminish potential antigen escape in these high-risk leukemias. Our preclinical data credential FLT3CART as a highly effective immunotherapeutic strategy for both FLT3- mutant AML and KMT2A-rearranged ALL which is poised for further investigation and clinical translation.
- Published
- 2022
- Full Text
- View/download PDF
3. Therapeutic potential of ruxolitinib and ponatinib in patients with EPOR-rearranged Philadelphia chromosome-like acute lymphoblastic leukemia
- Author
-
Lisa M. Niswander, Joseph P. Loftus, Élodie Lainey, Aurélie Caye-Eude, Morgane Pondrom, David A. Hottman, Ilaria Iacobucci, Charles G. Mullighan, Nitin Jain, Marina Konopleva, Hélène Cavé, André Baruchel, Pierre S. Rohrlich, and Sarah K. Tasian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
4. Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia
- Author
-
Joseph P. Loftus, Anella Yahiaoui, Patrick A. Brown, Lisa M. Niswander, Asen Bagashev, Min Wang, Allyson Schauf, Stacey Tannheimer, and Sarah K. Tasian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Survival of infants with KMT2A-rearranged (R) acute lymphoblastic leukemia (ALL) remains dismal despite intensive chemotherapy. We observed constitutive phosphorylation of spleen tyrosine kinase (SYK) and associated signaling proteins in infant ALL patient-derived xenograft (PDX) model specimens and hypothesized that the SYK inhibitor entospletinib would inhibit signaling and cell growth in vitro and leukemia proliferation in vivo. We further predicted that combined entospletinib and chemotherapy could augment anti-leukemia effects. Basal kinase signaling activation and HOXA9/MEIS1 expression differed among KMT2A-R (KMT2A-AFF1 [n=4], KMT2A-MLLT3 [n=1], KMT2A-MLLT1 [n=4]) and non-KMT2A-R [n=3] ALL specimens and stratified by genetic subgroup. Incubation of KMT2A-R ALL cells in vitro with entospletinib inhibited methylcellulose colony formation and SYK pathway signaling in a dose-dependent manner. In vivo inhibition of leukemia proliferation with entospletinib monotherapy was observed in RAS-wild-type KMT2A-AFF1, KMT2A-MLLT3, and KMT2A-MLLT1 ALL PDX models with enhanced activity in combination with vincristine chemotherapy in several models. Surprisingly, entospletinib did not decrease leukemia burden in two KMT2A-AFF1 PDX models with NRAS/ or KRAS mutations, suggesting potential RAS-mediated resistance to SYK inhibition. As hypothesized, superior inhibition of ALL proliferation was observed in KMT2A-AFF1 PDX models treated with entospletinib and the MEK inhibitor selumetinib versus vehicle or inhibitor monotherapies (p
- Published
- 2020
- Full Text
- View/download PDF
5. Systematic preclinical evaluation of CD33-directed chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia defines optimized construct design
- Author
-
Haiying Qin, Nirali N Shah, Terry J Fry, Lila Yang, John A Chukinas, Samiksha Tarun, Marie Pouzolles, Christopher D Chien, Lisa M Niswander, Anthony R Welch, Sarah K Tasian, and Naomi A Taylor
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
6. Therapeutic potential of ruxolitinib and ponatinib in patients with EPOR-rearranged Philadelphia chromosome-like acute lymphoblastic leukemia
- Author
-
Morgane Pondrom, Nitin Jain, Marina Konopleva, Sarah K. Tasian, André Baruchel, Aurélie Caye-Eude, Joseph P. Loftus, Ilaria Iacobucci, Lisa M Niswander, Pierre Rohrlich, Charles G. Mullighan, David A Hottman, Elodie Lainey, and Hélène Cavé
- Subjects
Oncology ,medicine.medical_specialty ,Ruxolitinib ,business.industry ,Lymphoblastic Leukemia ,Ponatinib ,Hematology ,Philadelphia chromosome ,medicine.disease ,Erythropoietin receptor ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Medicine ,In patient ,business ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
7. Clinical Experience with Azacitidine and Venetoclax +/- Gemtuzumab in Pediatric Patients with Relapsed or Refractory Acute Leukemias: A Report from the Children's Hospital of Philadelphia
- Author
-
Lisa M Niswander, Perry Chung, Caroline Diorio, and Sarah K Tasian
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
8. Preclinical In Vivo Activity of the Menin Inhibitor Ziftomenib (KO-539) in Pediatric KMT2A-Rearranged Acute Lymphoblastic Leukemia
- Author
-
Catherine D Falkenstein, Lisa M Niswander, Linda Kessler, Blake Tomkinson, Francis Burrows, and Sarah K Tasian
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
9. Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia
- Author
-
Min Wang, Joseph P. Loftus, Lisa M Niswander, Stacey Tannheimer, Patrick A. Brown, Anella Yahiaoui, Sarah K. Tasian, Asen Bagashev, and Allyson Schauf
- Subjects
Vincristine ,Indazoles ,Syk ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,biology ,business.industry ,MEK inhibitor ,Infant ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,KMT2A ,Pyrazines ,biology.protein ,Selumetinib ,Cancer research ,Heterografts ,business ,Myeloid-Lymphoid Leukemia Protein ,030215 immunology ,medicine.drug - Abstract
Survival of infants with KMT2A-rearranged acute lymphoblastic leukemia (ALL) remains dismal despite intensive chemotherapy. We observed constitutive phosphorylation of spleen tyrosine kinase (SYK) and associated signaling proteins in infant ALL patient-derived xenograft (PDX) model specimens and hypothesized that the SYK inhibitor entospletinib would inhibit signaling and cell growth in vitro and leukemia proliferation in vivo. We further predicted that combined entospletinib and chemotherapy could augment anti-leukemia effects. Basal kinase signaling activation and HOXA9/MEIS1 expression differed among KMT2Arearranged (KMT2A-AFF1 [n=4], KMT2A-MLLT3 [n=1], KMT2A-MLLT1 [n=4]) and non-KMT2A-rearranged [n=3] ALL specimens and stratified by genetic subgroup. Incubation of KMT2A-rearranged ALL cells in vitro with entospletinib inhibited methylcellulose colony formation and SYK pathway signaling in a dose-dependent manner. In vivo inhibition of leukemia proliferation with entospletinib monotherapy was observed in RAS-wild-type KMT2A-AFF1, KMT2A-MLLT3, and KMT2A-MLLT1 ALL PDX models with enhanced activity in combination with vincristine chemotherapy in several models. Surprisingly, entospletinib did not decrease leukemia burden in two KMT2A-AFF1 PDX models with NRAS or KRAS mutations, suggesting potential RAS-mediated resistance to SYK inhibition. As hypothesized, superior inhibition of ALL proliferation was observed in KMT2A-AFF1 PDX models treated with entospletinib and the MEK inhibitor selumetinib versus vehicle or inhibitor monotherapies (P
- Published
- 2020
10. Systematic preclinical evaluation of CD33-directed chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia defines optimized construct design
- Author
-
Naomi Taylor, Terry J. Fry, Christopher D. Chien, Lila Yang, Sarah K. Tasian, Nirali N. Shah, Haiying Qin, Marie Pouzolles, John Chukinas, Samiksha Tarun, Anthony R. Welch, and Lisa M Niswander
- Subjects
Male ,Cancer Research ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,Sialic Acid Binding Ig-like Lectin 3 ,Immunology ,CD33 ,Immunotherapy, Adoptive ,CD19 ,Mice ,medicine ,Animals ,Humans ,Immunology and Allergy ,RC254-282 ,Pharmacology ,Receptors, Chimeric Antigen ,biology ,business.industry ,Correction ,CD28 ,Myeloid leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Molecular Medicine ,Female ,business - Abstract
BackgroundSuccessful development of chimeric antigen receptor (CAR) T cell immunotherapy for children and adults with relapsed/refractory acute myeloid leukemia (AML) is highly desired given their poor clinical prognosis and frequent inability to achieve cure with conventional chemotherapy. Initial experiences with CD19 CAR T cell immunotherapy for patients with B-cell malignancies highlighted the critical impact of intracellular costimulatory domain selection (CD28 vs 4-1BB (CD137)) on CAR T cell expansion and in vivo persistence that may impact clinical outcomes. However, the impact of costimulatory domains on the efficacy of myeloid antigen-directed CAR T cell immunotherapy remains unknown.MethodsIn this preclinical study, we developed six CAR constructs targeting CD33, a highly expressed and validated AML target, comprised of one of three single-chain variable fragments with CD3ζ and either CD28 or 4-1BB costimulatory domains. We systematically compared the preclinical in vitro and in vivo efficacy of T cells lentivirally transduced with CD33 CAR constructs (CD33CARTs) against human AML.ResultsWe observed potent in vitro cytokine production and cytotoxicity of CD33CARTs incubated with human CD33+ AML cell lines, as well as robust in vivo antileukemia activity in cell line and childhood AML patient-derived xenograft (PDX) models. Gemtuzumab-based CD33CARTs were unexpectedly toxic in vivo in animal models despite observed in vitro anti-leukemia activity. CD28-based CD33CARTs consistently induced more robust inhibition of leukemia proliferation in AML cell line and PDX models than did 4-1BB-based CD33CARTs. A ‘best-in-class’ lintuzumab-CD28/CD3ζ CAR construct was thus selected for clinical translation.ConclusionsCD33 is a critical antigen for potential immunotherapeutic targeting in patients with AML. Based on this rigorous preclinical evaluation, our validated clinical grade lintuzumab-CD28/CD3ζ CD33CART immunotherapy is now under evaluation in a first-in-child/first-in-human phase 1 clinical trial for children and adolescents/young adults with relapsed/refractory AML.Trial registration numberclinicaltrials.gov;NCT03971799.
- Published
- 2021
11. Long-acting PGE2 and Lisinopril Mitigate H-ARS
- Author
-
Laura M. Calvi, Lisa M. Niswander, Seana C. Catherman, Kathleen E. McGrath, Jerry Saunders, Craig N. Morrell, Sara Ture, Meetha Medhora, James Palis, Paul D. Kingsley, Anne D. Koniski, and Jacqueline P. Williams
- Subjects
Blood Platelets ,Combination therapy ,Platelet Aggregation ,Biophysics ,Drug Evaluation, Preclinical ,Angiotensin-Converting Enzyme Inhibitors ,Pharmacology ,Hemorrhagic Disorders ,Platelet Factor 4 ,Article ,Thrombopoiesis ,Mice ,In vivo ,Bone Marrow ,Lisinopril ,16,16-Dimethylprostaglandin E2 ,von Willebrand Factor ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Platelet ,Prostaglandin E2 ,Radiation ,business.industry ,Endothelial Cells ,Thrombocytopenia ,In vitro ,Mice, Inbred C57BL ,Haematopoiesis ,P-Selectin ,Radiation Injuries, Experimental ,C-Reactive Protein ,Acute Radiation Syndrome ,Cesium Radioisotopes ,Gamma Rays ,Female ,Endothelium, Vascular ,business ,Megakaryocytes ,Whole-Body Irradiation ,medicine.drug - Abstract
Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E(2) (dmPGE(2)), a longer-acting analogue of PGE(2), promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE(2) and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE(2) and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE(2) and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE(2), alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin(−)CD45(−)CD31(+)Sca-1(−) sinusoidal endothelial cells, while combined dmPGE(2) and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE(2) and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.
- Published
- 2020
12. Multi-Modal Targeting of FLT3 with Chimeric Antigen Receptor T Cell Immunotherapy and Tyrosine Kinase Inhibition in High-Risk Pediatric Leukemias
- Author
-
Zachary Graff, Lillie Leach, Asen Bagashev, Sarah K. Tasian, Lisa M Niswander, and Terry J. Fry
- Subjects
Chemistry ,hemic and lymphatic diseases ,Immunology ,Cancer research ,Cell Biology ,Hematology ,Biochemistry ,T cell immunotherapy ,Tyrosine kinase ,Chimeric antigen receptor - Abstract
Background: Clinical outcomes for children with FLT3-mutant AML and infants with KMT2A-rearranged (KMT2A-R) B-ALL remain dismal. These leukemias share a common feature of aberrant activation of FLT3 kinase signaling, which occurs by activating FLT3 mutations in AML and by overexpression of wild-type FLT3 in KMT2A-R ALL. Several FLT3 tyrosine kinase inhibitors (FLT3i) are approved for adults with FLT3-mutant AML, but potential efficacy against KMT2A-R ALL remains incompletely characterized and may differ from responses in AML. We previously developed and preclinically validated chimeric antigen receptor (CAR) T cells directed against FLT3 (FLT3CART), which importantly showed potent anti-leukemia activity in preclinical models of both childhood FLT3-mutant AML and infant KMT2A-R ALL (Chien CD et al. ASH 2016). In the current studies, we hypothesized that combinatorial targeting of these two high-risk leukemia subtypes with FLT3CART and the selective next-generation FLT3i gilteritinib would have superior activity and potentially mitigate therapeutic resistance now known to occur with kinase inhibitors or CAR T cell immunotherapy. Methods and Results: We first assessed in vitro sensitivity of human FLT3-mutant AML and KMT2A-R ALL cell lines to gilteritinib, a second-generation selective FLT3i with established clinical activity in FLT3-mutant AML and unknown activity in KMT2A-R ALL. As detrimental effects of kinase inhibitors (e.g., dasatinib, ruxolitinib) upon CAR T cells have been reported, we evaluated for similar effects with gilteritinib co-incubated in vitro with CD3/CD28-bead activated healthy human donor T cells. However, we observed minimal deleterious effects of gilteritinib on normal T cell viability, immunophenotype, and IL-2 and interferon-gamma (IFNg) production. We validated combinatorial effects of gilteritinib and FLT3CART-induced cytotoxicity against FLT3-mutant AML and KMT2A-R ALL cell lines in vitro without impairment of IL-2/IFNg production. We then assessed this dual therapy approach in luciferase+ FLT3-mutant AML (MOLM14) and KMT2A-R ALL (SEM) cell line murine xenograft models. As predicted, both FLT3CART and gilteritinib monotherapies transiently inhibited in vivo leukemia proliferation, although leukemia progression eventually occurred. Conversely, FLT3CART and gilteritinib combination therapy strikingly induced enhanced and sustained leukemia clearance in all assessed AML and ALL cell line xenograft models (Figure 1). Confirmatory studies in our established childhood FLT3-mutant AML and KMT2A-R ALL patient-derived xenograft (PDX) models have also demonstrated potent anti-leukemia efficacy of combined FLT3CART and gilteritinib therapy. Earlier-generation FLT3i have been reported to increase cell surface FLT3 expression on FLT3-mutant AML cells. Given the known importance of target antigen site density for CAR T cell efficacy, we reasoned that a sequential approach to dual therapy with FLT3i 'priming' followed by FLT3CART may be superior to a simultaneous treatment strategy. In vitro studies with leukemia cell lines and in vivo studies with PDX models indeed confirmed gilteritinib-induced increases in FLT3 surface antigen density in FLT3-mutant AML cells. Intriguingly, we observed contrasting effects in KMT2A-R ALL cell lines and PDX with decreased surface FLT3 expression upon gilteritinib exposure. Ongoing studies are currently validating gilteritinib priming for FLT3CART given these initial data suggesting potentially divergent sequencing approaches in FLT3-mutant AML versus KMT2A-R ALL. Conclusions: Taken together, our preclinical studies demonstrate that dual targeting with FLT3CART immunotherapy and gilteritinib is a promising therapeutic strategy in FLT3-mutant AML and, importantly, also in KMT2A-R ALL. Notably, we also report minimal negative effects of gilteritinib on FLT3CART, suggesting that FLT3i may be used to enhance CAR T cell immunotherapy without inhibiting T cell function. Phase 1 clinical trials of FLT3CART will open soon for adults and children with FLT3-mutant AML and/or KMT2A-R ALL. Figure 1 Figure 1. Disclosures Fry: Sana Biotechnology: Current Employment, Current equity holder in publicly-traded company; ElevateBio: Research Funding. Tasian: Kura Oncology: Consultancy; Aleta Biotherapeutics: Consultancy; Gilead Sciences: Research Funding; Incyte Corporation: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
13. Precision Co-Targeting of the Thymic Stromal Lymphopoietin Receptor in Childhood CRLF2-Rearranged Acute Lymphoblastic Leukemia
- Author
-
Haiying Qin, Sarah K. Tasian, Savannah Ross, Lisa M Niswander, Asen Bagashev, Terry J. Fry, and Joseph P. Loftus
- Subjects
business.industry ,Lymphoblastic Leukemia ,Immunology ,Cancer research ,Medicine ,THYMIC STROMAL LYMPHOPOIETIN RECEPTOR ,Cell Biology ,Hematology ,business ,Biochemistry - Abstract
Introduction : Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is associated with high rates of chemoresistance and relapse. CRLF2 (cytokine receptor-like factor 2) rearrangements occur in 50% of Ph-like and 60% of Down Syndrome (DS)-associated ALL and induce constitutive JAK/STAT and other kinase signaling. Current clinical trials are studying chemotherapy with the JAK inhibitor ruxolitinib in patients with CRLF2-rearranged Ph-like ALL, but results are not yet known. While chimeric antigen receptor T-cell (CART) immunotherapies have induced remarkable remissions in children with relapsed/refractory B-ALL, approximately 50% of CD19CART-treated patients will relapse again, many with CD19 antigen loss. New therapies are needed to prevent relapse and overcome immunotherapeutic resistance. Methods : We previously developed CAR T cells targeting the thymic stromal lymphopoietin receptor (TSLPR; encoded by CRLF2) and demonstrated potent preclinical activity in Ph-like ALL models (Qin Blood 2015), which has led to a soon-to-open phase 1 clinical trial for patients with relapsed/refractory CRLF2-overexpressing ALL. In the current preclinical studies, we hypothesized that combinatorial targeting with bispecific TSLPRxCD19CART or TSLPRxCD22CART (Ross Cancer Res 2020) or with TSLPRCART + ruxolitinib will have superior activity against CRLF2-rearranged Ph-like and DS-ALL. Results : TSLPRCART treatment of CRLF2-rearranged ALL cell line (n=1) and patient-derived xenograft (PDX) models potently inhibited leukemia proliferation in vitro and in vivo and induced long-term 'cure' of xenograft mice. However, co-administration of TSLPRCART + ruxolitinib markedly diminished in vivo T cell numbers, blunted cytokine production, and facilitated leukemia relapse, which could be abrogated by delaying ruxolitinib. Importantly, ruxolitinib co-treatment prevented severe TSLPRCART-induced cytokine release syndrome (CRS) and animal death. Interestingly, ruxolitinib withdrawal led to return of T-cell functionality with re-detection of TSLPRCART in peripheral blood, induction of IFN-γ production, and leukemia clearance upon CRLF2+ ALL rechallenge (Figure 1). Conclusions: In these preclinical studies, we report potent activity of TSLPRCART in cell line (n=1) and PDX models of childhood CRLF2-rearranged Ph-like ALL (n=2) and DS-ALL (n=2) and, interestingly, deleterious effects of concomitant JAK inhibition upon CAR T cell functionality. We demonstrated that ruxolitinib co-administration impaired in vivo TSLPRCART-induced ALL cell killing but was also beneficial in protection against life-threatening cytokine release syndrome in co-treated animals. Importantly, TSLPRCART was not eliminated, only suppressed, by JAKi co-treatment with restoration of T cell functionality upon ruxolitinib removal and/or leukemia relapse/rechallenge studies. Ongoing studies are defining optimal TSLPRCART + ruxolitinib sequence(s) to maximize both anti-leukemia efficacy and potential CRS mitigation, as well as assessing in vivo efficacy of bispecific TSLPRCARTs in CRLF2-R Ph-like ALL and DS-ALL PDX models for future translation and clinical evaluation in next-generation trials. Figure 1 Figure 1. Disclosures Fry: ElevateBio: Research Funding; Sana Biotechnology: Current Employment, Current equity holder in publicly-traded company. Tasian: Aleta Biotherapeutics: Consultancy; Kura Oncology: Consultancy; Gilead Sciences: Research Funding; Incyte Corporation: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
14. Mutation in mouse hei10, an e3 ubiquitin ligase, disrupts meiotic crossing over.
- Author
-
Jeremy O Ward, Laura G Reinholdt, William W Motley, Lisa M Niswander, Dekker C Deacon, Laurie B Griffin, Kristofor K Langlais, Vickie L Backus, Kerry J Schimenti, Marilyn J O'Brien, John J Eppig, and John C Schimenti
- Subjects
Genetics ,QH426-470 - Abstract
Crossing over during meiotic prophase I is required for sexual reproduction in mice and contributes to genome-wide genetic diversity. Here we report on the characterization of an N-ethyl-N-nitrosourea-induced, recessive allele called mei4, which causes sterility in both sexes owing to meiotic defects. In mutant spermatocytes, chromosomes fail to congress properly at the metaphase plate, leading to arrest and apoptosis before the first meiotic division. Mutant oocytes have a similar chromosomal phenotype but in vitro can undergo meiotic divisions and fertilization before arresting. During late meiotic prophase in mei4 mutant males, absence of cyclin dependent kinase 2 and mismatch repair protein association from chromosome cores is correlated with the premature separation of bivalents at diplonema owing to lack of chiasmata. We have identified the causative mutation, a transversion in the 5' splice donor site of exon 1 in the mouse ortholog of Human Enhancer of Invasion 10 (Hei10; also known as Gm288 in mouse and CCNB1IP1 in human), a putative B-type cyclin E3 ubiquitin ligase. Importantly, orthologs of Hei10 are found exclusively in deuterostomes and not in more ancestral protostomes such as yeast, worms, or flies. The cloning and characterization of the mei4 allele of Hei10 demonstrates a novel link between cell cycle regulation and mismatch repair during prophase I.
- Published
- 2007
- Full Text
- View/download PDF
15. A mammal-specific Doublesex homolog associates with male sex chromatin and is required for male meiosis.
- Author
-
Shinseog Kim, Satoshi H Namekawa, Lisa M Niswander, Jeremy O Ward, Jeannie T Lee, Vivian J Bardwell, and David Zarkower
- Subjects
Genetics ,QH426-470 - Abstract
Gametogenesis is a sexually dimorphic process requiring profound differences in germ cell differentiation between the sexes. In mammals, the presence of heteromorphic sex chromosomes in males creates additional sex-specific challenges, including incomplete X and Y pairing during meiotic prophase. This triggers formation of a heterochromatin domain, the XY body. The XY body disassembles after prophase, but specialized sex chromatin persists, with further modification, through meiosis. Here, we investigate the function of DMRT7, a mammal-specific protein related to the invertebrate sexual regulators Doublesex and MAB-3. We find that DMRT7 preferentially localizes to the XY body in the pachytene stage of meiotic prophase and is required for male meiosis. In Dmrt7 mutants, meiotic pairing and recombination appear normal, and a transcriptionally silenced XY body with appropriate chromatin marks is formed, but most germ cells undergo apoptosis during pachynema. A minority of mutant cells can progress to diplonema, but many of these escaping cells have abnormal sex chromatin lacking histone H3K9 di- and trimethylation and heterochromatin protein 1beta accumulation, modifications that normally occur between pachynema and diplonema. Based on the localization of DMRT7 to the XY body and the sex chromatin defects observed in Dmrt7 mutants, we conclude that DMRT7 plays a role in the sex chromatin transformation that occurs between pachynema and diplonema. We suggest that DMRT7 may help control the transition from meiotic sex chromosome inactivation to postmeiotic sex chromatin in males. In addition, because it is found in all branches of mammals, but not in other vertebrates, Dmrt7 may shed light on evolution of meiosis and of sex chromatin.
- Published
- 2007
- Full Text
- View/download PDF
16. SDF-1 dynamically mediates megakaryocyte niche occupancy and thrombopoiesis at steady state and following radiation injury
- Author
-
Paul D. Kingsley, Katherine H. Fegan, Kathleen E. McGrath, Lisa M. Niswander, and James Palis
- Subjects
Stromal cell ,Megakaryocyte Progenitor Cells ,Immunology ,Chemotaxis ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,biology.protein ,Stromal cell-derived factor 1 ,Bone marrow ,Thrombopoiesis ,Progenitor - Abstract
Megakaryocyte (MK) development in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine stromal cell-derived factor-1 (SDF-1), signaling through CXCR4, is implicated in the maturational chemotaxis of MKs toward sinusoidal vessels. Here, we demonstrate that both IV administration of SDF-1 and stabilization of endogenous SDF-1 acutely increase MK-vasculature association and thrombopoiesis with no change in MK number. In the setting of radiation injury, we find dynamic fluctuations in marrow SDF-1 distribution that spatially and temporally correlate with variations in MK niche occupancy. Stabilization of altered SDF-1 gradients directly affects MK location. Importantly, these SDF-1-mediated changes have functional consequences for platelet production, as the movement of MKs away from the vasculature decreases circulating platelets, while MK association with the vasculature increases circulating platelets. Finally, we demonstrate that manipulation of SDF-1 gradients can improve radiation-induced thrombocytopenia in a manner additive with earlier TPO treatment. Taken together, our data support the concept that SDF-1 regulates the spatial distribution of MKs in the marrow and consequently circulating platelet numbers. This knowledge of the microenvironmental regulation of the MK lineage could lead to improved therapeutic strategies for thrombocytopenia.
- Published
- 2014
- Full Text
- View/download PDF
17. Improved quantitative analysis of primary bone marrow megakaryocytes utilizing imaging flow cytometry
- Author
-
Kathleen E. McGrath, James Palis, Lisa M. Niswander, and John C. Kennedy
- Subjects
Imaging flow cytometry ,Histology ,medicine.diagnostic_test ,urogenital system ,Cell Biology ,Biology ,Pathology and Forensic Medicine ,Flow cytometry ,Cell biology ,Primary bone ,medicine.anatomical_structure ,Megakaryocyte ,Immunology ,medicine ,Platelet ,Bone marrow ,Cytometry ,Megakaryopoiesis - Abstract
Life-threatening thrombocytopenia can develop following bone marrow injury due to decreased platelet production from megakaryocytes (MKs). However, the study of primary MKs has been complicated by their low frequency in the bone marrow and by technical challenges presented by their unique maturation properties. More accurate and efficient methods for the analysis of in vivo MKs are needed to enhance our understanding of megakaryopoiesis and ultimately develop new therapeutic strategies for thrombocytopenia. Imaging flow cytometry (IFC) combines the morphometric capabilities of microscopy with the high-throughput analyses of flow cytometry (FC). Here, we investigate the application of IFC on the ImageStreamX platform to the analysis of primary MKs isolated from murine bone marrow. Our data highlight and address technical challenges for conventional FC posed by the wide range of cellular size within the MK lineage as well as the shared surface phenotype with abundant platelet progeny. We further demonstrate that IFC can be used to reproducibly and efficiently quantify the frequency of primary murine MKs in the marrow, both at steady-state and in the setting of radiation-induced bone marrow injury, as well as assess their ploidy distribution. The ability to accurately analyze the full spectrum of maturing MKs in the bone marrow now allows for many possible applications of IFC to enhance our understanding of megakaryopoiesis and platelet production. © 2014 International Society for Advancement of Cytometry
- Published
- 2014
- Full Text
- View/download PDF
18. Imaging Flow Cytometric Analysis of Primary Bone Marrow Megakaryocytes
- Author
-
Lisa M, Niswander, James, Palis, and Kathleen E, McGrath
- Subjects
Blood Platelets ,Mice ,Animals ,Bone Marrow Cells ,Flow Cytometry ,Megakaryocytes ,Thrombocytopenia - Abstract
In light of the indispensible role of platelets in the maintenance of hemostasis, understanding the biology of platelet production from bone marrow megakaryocytes (MKs) may uncover new therapeutic strategies for thrombocytopenia. While there has been much recent interest in optimizing culture systems to facilitate the study of the morphologically unique MK lineage, these systems lack the intricacy of in vivo megakaryopoiesis. Given the limitations of many common techniques for the in vivo study of MKs, in this chapter we describe a method to quantify and analyze primary murine bone marrow megakaryocytes utilizing imaging flow cytometry.
- Published
- 2016
19. Imaging Flow Cytometric Analysis of Primary Bone Marrow Megakaryocytes
- Author
-
Kathleen E. McGrath, James Palis, and Lisa M. Niswander
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,Biology ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Primary bone ,Megakaryocyte ,In vivo ,Hemostasis ,medicine ,Platelet ,Bone marrow ,Megakaryopoiesis - Abstract
In light of the indispensible role of platelets in the maintenance of hemostasis, understanding the biology of platelet production from bone marrow megakaryocytes (MKs) may uncover new therapeutic strategies for thrombocytopenia. While there has been much recent interest in optimizing culture systems to facilitate the study of the morphologically unique MK lineage, these systems lack the intricacy of in vivo megakaryopoiesis. Given the limitations of many common techniques for the in vivo study of MKs, in this chapter we describe a method to quantify and analyze primary murine bone marrow megakaryocytes utilizing imaging flow cytometry.
- Published
- 2015
- Full Text
- View/download PDF
20. End-of-life care for children enrolled in a community-based pediatric palliative care program
- Author
-
Philene Cromwell, Alyssa Gupton, Lisa M. Niswander, Jeanne Chirico, and David N. Korones
- Subjects
Pediatrics ,medicine.medical_specialty ,Adolescent ,MEDLINE ,New York ,Community Networks ,Young Adult ,medicine ,Humans ,Young adult ,Child ,General Nursing ,Retrospective Studies ,Community based ,Terminal Care ,business.industry ,Palliative Care ,Case manager ,Infant ,Retrospective cohort study ,General Medicine ,Home Care Services ,Pediatric palliative care ,Anesthesiology and Pain Medicine ,Home visits ,Family medicine ,Child, Preschool ,Organizational Case Studies ,business ,End-of-life care - Abstract
Despite recent strides in pediatric palliative care (PPC), there are few published data on community-based care of dying children.Our aim was to describe end-of-life care during the last 6 months of life for children on a community-based PPC program.We conducted a retrospective review of children21 years old who died while enrolled in a community-based PPC program (CompassionNet) from December 2004 through May 2008. Data were extracted on the last 6 months of life from hospital records (University of Rochester) and records of CompassionNet and its home-nursing team.Thirty-six children died while enrolled in the CompassionNet program; diagnoses included cancer 20 (56%), genetic/neurologic disorders 7 (19%), cardiac anomalies 6 (17%), and other 3 (8%). In the last 6 months of life, there were a median of 2 emergency room visits (range 0-12) and 2 unplanned hospitalizations (range 0-6). Home visits by CompassionNet (case manager, physician, nurse) ranged from 1 to 121 (median 24). A "goals of care" discussion was documented for 29 children (81%), occurring a median of 16 days before death (range 0-118). Sixteen children died at home (44%); 20 died in the hospital (56%). Of the 16 families with a documented preference for location of death, 14 children died in this location (88%). Thirty-two families (89%) had bereavement support through CompassionNet.Children who die of complex chronic conditions spend the majority of their last 6 months of life at home. Community-based PPC can contribute substantially to their care and comfort.
- Published
- 2014
21. Improved quantitative analysis of primary bone marrow megakaryocytes utilizing imaging flow cytometry
- Author
-
Lisa M, Niswander, Kathleen E, McGrath, John C, Kennedy, and James, Palis
- Subjects
Mice, Inbred C57BL ,Mice ,Animals ,Female ,Flow Cytometry ,Megakaryocytes - Abstract
Life-threatening thrombocytopenia can develop following bone marrow injury due to decreased platelet production from megakaryocytes (MKs). However, the study of primary MKs has been complicated by their low frequency in the bone marrow and by technical challenges presented by their unique maturation properties. More accurate and efficient methods for the analysis of in vivo MKs are needed to enhance our understanding of megakaryopoiesis and ultimately develop new therapeutic strategies for thrombocytopenia. Imaging flow cytometry (IFC) combines the morphometric capabilities of microscopy with the high-throughput analyses of flow cytometry (FC). Here, we investigate the application of IFC on the ImageStream(X) platform to the analysis of primary MKs isolated from murine bone marrow. Our data highlight and address technical challenges for conventional FC posed by the wide range of cellular size within the MK lineage as well as the shared surface phenotype with abundant platelet progeny. We further demonstrate that IFC can be used to reproducibly and efficiently quantify the frequency of primary murine MKs in the marrow, both at steady-state and in the setting of radiation-induced bone marrow injury, as well as assess their ploidy distribution. The ability to accurately analyze the full spectrum of maturing MKs in the bone marrow now allows for many possible applications of IFC to enhance our understanding of megakaryopoiesis and platelet production.
- Published
- 2013
22. GPCR/EGFR cross talk is conserved in gonadal and adrenal steroidogenesis but is uniquely regulated by matrix metalloproteinases 2 and 9 in the ovary
- Author
-
Liliana Carbajal, Lisa M. Niswander, Anindita Biswas, Hen Prizant, and Stephen R. Hammes
- Subjects
MAPK/ERK pathway ,endocrine system ,medicine.medical_specialty ,Gonadotropins, Equine ,Ovary ,Adrenocorticotropic hormone ,Biology ,Matrix Metalloproteinase Inhibitors ,Mice ,Endocrinology ,Adrenocorticotropic Hormone ,Ovarian Follicle ,Epidermal growth factor ,Internal medicine ,Adrenal Glands ,Nitriles ,medicine ,Butadienes ,Animals ,Humans ,Ovarian follicle ,Receptor ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Progesterone ,G protein-coupled receptor ,Original Research ,General Medicine ,Receptor Cross-Talk ,Luteinizing Hormone ,Receptors, LH ,Tyrphostins ,Cyclic AMP-Dependent Protein Kinases ,ErbB Receptors ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Receptors, Corticotropin ,Doxycycline ,Quinazolines ,Matrix Metalloproteinase 2 ,Female ,Signal transduction ,Signal Transduction - Abstract
Previous work has demonstrated that cross talk between G protein-coupled LH receptors and epidermal growth factor receptors (EGFR) is essential for LH-induced steroid production in ovarian follicles and testicular Leydig cells. Here we demonstrate that G protein-coupled receptor (GPCR)/EGFR cross talk is also required for ACTH-induced steroidogenesis in Y1 adrenal cells. Moreover, we confirm that the signaling pathway from GPCR to Erk activation is conserved in all three steroidogenic tissues. ACTH or LH induces Gα(s), resulting in elevated cAMP and protein kinase A activation. cAMP/protein kinase A then triggers EGFR trans-activation, which promotes Erk signaling and subsequent steroidogenesis. Interestingly, although EGFR trans-activation is conserved in all three tissues, the specific mechanisms regulating this receptor cross talk differ. ACTH and LH trigger matrix metalloproteinase (MMP)-mediated release of EGFR ligands in adrenal and gonadal cells, respectively. However, this extracellular, ligand-dependent EGFR transactivation is required only for LH-induced steroidogenesis in ovarian follicles, reflecting the unique requirement of cell-cell cross talk for ovarian steroid production. Furthermore, MMP2 and MMP9 appear to regulate LH-induced steroidogenesis in mouse ovarian follicles, because a specific MMP2/9 inhibitor as well as the MMP2/9 inhibitor doxycycline suppress LH-induced follicular steroid production in vitro. Notably, although EGFR or MMP inhibition minimally affects estrous cycling in female mice, they attenuate ovarian steroidogenesis in response to LHR overstimulation in vivo. These results may have implications with regard to EGFR inhibitor use in various cancers as well as in polycystic ovarian syndrome, where excess LH-driven ovarian androgen production might be controlled by MMP2/9 inhibition.
- Published
- 2011
23. Stratifying osteosarcoma: minimizing and maximizing therapy
- Author
-
Lisa M. Niswander and Su Young Kim
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Integrins ,Bevacizumab ,medicine.medical_treatment ,MEDLINE ,Disease ,Antibodies, Monoclonal, Humanized ,Article ,Targeted therapy ,medicine ,Temozolomide ,Humans ,Survivors ,Intensive care medicine ,Osteosarcoma ,business.industry ,Cancer ,Antibodies, Monoclonal ,medicine.disease ,Surgery ,Dacarbazine ,Receptors, Vascular Endothelial Growth Factor ,Treatment Outcome ,Oncology ,Molecular targets ,Quinazolines ,business ,medicine.drug - Abstract
Patients who are newly diagnosed with osteosarcoma face a daunting year of medical and surgical therapy, often filled with hospitalizations and changes in lifestyle. Fortunately, the majority of patients endure this struggle to become long-term survivors. However, follow-up studies of cancer survivors are revealing the sequelae of this curative therapy. Just as disturbingly, there remains a large subset of patients for whom conventional therapy is inadequate and who succumb to disease. In this review, we propose that therapeutic strategies for osteosarcoma patients must rely on stratification of patients into risk categories, in order to minimize therapy for some, while expanding treatment for others. We then focus on two molecular targets for the treatment of patients with high-risk osteosarcoma.
- Published
- 2010
24. Mutation in mouse hei10, an e3 ubiquitin ligase, disrupts meiotic crossing over
- Author
-
Marilyn J. O'Brien, Laura G. Reinholdt, Vickie L. Backus, Lisa M Niswander, Laurie B. Griffin, William W. Motley, Kerry J. Schimenti, Dekker C. Deacon, Jeremy O. Ward, Kristofor K. Langlais, John J. Eppig, and John C. Schimenti
- Subjects
Male ,Cancer Research ,Base Pair Mismatch ,Eukaryotes ,Cell Cycle Proteins ,QH426-470 ,medicine.disease_cause ,Chromosomal crossover ,Mice ,Crossing Over, Genetic ,Meiotic Prophase I ,Genetics (clinical) ,Genetics ,Mice, Knockout ,Recombination, Genetic ,Mammals ,0303 health sciences ,Mutation ,Mice, Inbred C3H ,030302 biochemistry & molecular biology ,Mus (Mouse) ,3. Good health ,Ubiquitin ligase ,Vertebrates ,Female ,Research Article ,DNA repair ,Ubiquitin-Protein Ligases ,Biology ,03 medical and health sciences ,Prophase ,Meiosis ,medicine ,Animals ,Humans ,Molecular Biology ,Metaphase ,Ecology, Evolution, Behavior and Systematics ,Alleles ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Cyclin-Dependent Kinase 2 ,Genetics and Genomics ,Cell Biology ,Mice, Inbred C57BL ,biology.protein ,Cattle - Abstract
Crossing over during meiotic prophase I is required for sexual reproduction in mice and contributes to genome-wide genetic diversity. Here we report on the characterization of an N-ethyl-N-nitrosourea-induced, recessive allele called mei4, which causes sterility in both sexes owing to meiotic defects. In mutant spermatocytes, chromosomes fail to congress properly at the metaphase plate, leading to arrest and apoptosis before the first meiotic division. Mutant oocytes have a similar chromosomal phenotype but in vitro can undergo meiotic divisions and fertilization before arresting. During late meiotic prophase in mei4 mutant males, absence of cyclin dependent kinase 2 and mismatch repair protein association from chromosome cores is correlated with the premature separation of bivalents at diplonema owing to lack of chiasmata. We have identified the causative mutation, a transversion in the 5′ splice donor site of exon 1 in the mouse ortholog of Human Enhancer of Invasion 10 (Hei10; also known as Gm288 in mouse and CCNB1IP1 in human), a putative B-type cyclin E3 ubiquitin ligase. Importantly, orthologs of Hei10 are found exclusively in deuterostomes and not in more ancestral protostomes such as yeast, worms, or flies. The cloning and characterization of the mei4 allele of Hei10 demonstrates a novel link between cell cycle regulation and mismatch repair during prophase I., Author Summary Human infertility and reproductive complications have devastating social and monetary costs. Errors in meiosis during reproduction may lead to birth defects, spontaneous abortion, or infertility. Many of the genes essential for meiosis function in DNA repair and mutations in several of these genes have been shown to contribute to cancer. The identification of the genes necessary for normal meiosis is an important goal and will potentially influence the fields of reproductive and cancer biology. In this study, genetic screens in mice have generated the mutation mei4. mei4 causes male and female sterility by disrupting meiosis and altering the function of the DNA repair system known as mismatch repair. We have identified the causative mutation behind the mei4 phenotype in a gene called Human Enhancer of Invasion 10 or Hei10. This work demonstrates that Hei10 is essential for the completion of meiosis and that it functions to coordinate the DNA repair system and the progression of the cell cycle during meiosis.
- Published
- 2007
25. Prostaglandin E2 Promotes the Sequential Recovery of Bone Marrow Vasculature and the Megakaryocyte Lineage Following Radiation Injury
- Author
-
Katherine H. Fegan, Paul D. Kingsley, Laura M. Calvi, James Palis, Lisa M. Niswander, Seana C. Catherman, Andrew Seraichick, and Anne D. Koniski
- Subjects
Pathology ,medicine.medical_specialty ,Immunology ,Cell Biology ,Hematology ,Biology ,Total body irradiation ,Biochemistry ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,Cancer research ,Bone marrow ,Thrombopoiesis ,Progenitor cell ,Stem cell - Abstract
The sinusoidal vascular niche is the site of terminal megakaryopoiesis and platelet production in the bone marrow. The association of megakaryocytes (MKs) with the sinusoidal endothelium and microenvironmental cues from the vascular niche are critical for efficient thrombopoiesis. Thrombocytopenia is an important clinical problem that can cause significant morbidity for patients receiving genotoxic treatments such as chemotherapy and radiation therapy. Following myeloablative bone marrow injury, the recovery of both hematopoietic stem and progenitor cells (HSPCs) and the MK lineage is dependent on marrow vascular recovery (Hooper et al. Cell Stem Cell 2009, Avecilla et al. Nature 2004). Accordingly, we reasoned that improving recovery of the vasculature might serve as a rational therapeutic strategy for thrombocytopenia stemming from cytotoxic treatments. We focused our efforts on the arachidonic acid derivative prostaglandin E2 (PGE2), which regulates numerous physiologic processes including microvascular endothelial cell proliferation and angiogenesis. Within the context of the bone marrow, PGE2 also has diverse roles in HSPC expansion, survival, and trafficking. Importantly, systemic administration of the long-acting analogue 16,16-dimethyl PGE2 (dmPGE2) improves hematopoietic recovery and enhances overall survival following total body irradiation (TBI) in mice (Hoggatt et al. Blood Cells 2013). Intriguingly, following sublethal TBI, dmPGE2-induced recovery of peripheral platelets precedes restoration of red blood cells and neutrophils (Porter et al. Stem Cells 2013). We hypothesized that the preferential recovery of platelets following dmPGE2 administration is mediated by accelerated vascular recovery in the marrow. We tested this in a murine model of sublethal radiation injury in which we had previously characterized MK lineage kinetics following 4Gy TBI (Niswander et al. Blood 2014). First, the kinetics of MK progenitors (MKPs), maturing MKs, and platelets were determined at days 5, 7, and 10 in mice receiving 6mg/kg dmPGE2 or vehicle at 48 and 72 hours post-4Gy TBI. Mice receiving dmPGE2 had an early decrease in both MKPs and platelets, with no change in MKs at day 5 post-TBI. However, by day 10, dmPGE2-treated mice demonstrated significantly enhanced recovery of both MKs and circulating platelets compared to controls, but no difference in the number of immunophenotypic HSPCs. Having confirmed enhanced recovery of the MK lineage with dmPGE2 treatment, we next sought to characterize vascular recovery. Marked injury and dilation of the marrow sinusoidal vasculature occurs rapidly following TBI. Immunohistochemistry with morphometric analysis revealed no change in vascular dilation at day 5 post-TBI with dmPGE2 treatment. However, mice receiving dmPGE2 exhibited a more than 25% improvement in vascular dilation that was first evident at day 7 and was sustained at day 10 post-TBI. While vascular dilation post-TBI has been posited to correlate with loss of surrounding hematopoietic cells, improvement in vascular dilation with dmPGE2 treatment is contrastingly evident in the setting of slightly fewer (day 7) or equivalent (day 10) numbers of nucleated marrow cells compared to irradiated control mice. These data suggest that dmPGE2-mediated recovery of the vasculature coincides temporally with the accelerated onset of MK lineage recovery. In addition, immunohistochemical morphometric analyses indicate that dmPGE2-treated mice had an 80% increase in maturing MKs in physical contact with the vascular endothelium, the site of platelet production, at day 10 post-TBI. Taken together, these data support the concept that dmPGE2-mediated marrow vascular recovery following radiation injury enhances the recovery of the MK lineage and accelerates the production of circulating platelets. Modulation of vascular recovery with dmPGE2 may be a promising therapeutic strategy for thrombocytopenia secondary to myeloablative bone marrow injury. Disclosures Calvi: Fate Therapeutics: Patents & Royalties.
- Published
- 2015
- Full Text
- View/download PDF
26. Spatial and Temporal Fluctuations In Marrow SDF-1 Following Radiation Injury Regulate Megakaryocyte-Vascular Niche Interactions and Circulating Platelet Levels
- Author
-
James Palis, Paul D. Kingsley, Kathleen E. McGrath, Lisa M. Niswander, and Katherine H. Fegan
- Subjects
Endosteum ,Pathology ,medicine.medical_specialty ,Stromal cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,biology.protein ,Platelet ,Stromal cell-derived factor 1 ,Thrombopoiesis ,Bone marrow - Abstract
The development of megakaryocytes (MKs) in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine SDF-1 (CXCL12), signaling through receptor CXCR4, is produced by stromal cell populations throughout the marrow and is implicated in the maturational chemotaxis of MKs to the sinusoids. Understanding the regulation of MK localization has significance not only for optimal platelet production and the development of therapies for thrombocytopenia, but also in light of the recently proposed role for MKs in supporting hematopoietic stem cells (Heazlewood et al. 2013). In the injury setting of lethal total body irradiation (TBI), it was observed that radioresistant mature MKs relocate to the endosteal niche (Dominici et al. 2009, Olson et al. 2013). Complicating the study of marrow niches post-TBI is the vascular dilation that accompanies the drastic loss of marrow cells. Having confirmed that MKs relocate to the endosteum in our model of sublethal radiation-induced thrombocytopenia (4Gy TBI), we asked whether this localization is due to changes in the spatial distribution of the vasculature or to altered microenvironmental SDF-1. In agreement with other TBI models, we find a significant elevation in SDF-1 transcript levels in the marrow at days 1-3 following 4Gy TBI. Radioresistant MKs, which do not decrease in number until after 3 days, have significantly increased CXCR4 surface expression, a finding we also observe following SDF-1 stimulation of MKs both in vitro and in vivo. In situ hybridization was used to localize the spatial distribution of SDF-1 RNA in femoral marrow. At 2 days post-4Gy, a significant SDF-1 gradient develops with 30% higher SDF-1 message adjacent to the endosteum than in the central marrow. However, this gradient is dynamically eliminated 24 hours later at 3 days post-TBI. These shifts in SDF-1 expression are accompanied by parallel changes in the spatial distribution of MKs by immunohistochemistry. At 2 days post-TBI, there is over a 40% increase in MK in the endosteal niche. In contrast, MKs in the endosteal niche decrease by more than 15% at 3 days, coincident with a significant increase in the MKs associated with vascular endothelium. Thus, these data suggest that the spatial distribution of MKs is dependent upon the localization of SDF-1 in the rapidly fluctuating post-injury bone marrow. To determine if SDF-1 functionally contributes to MK niche changes, we stabilized endogenously-produced SDF-1 using Diprotin A, an inhibitor of SDF-1-inactivating protease DPP4. In uninjured marrow, Diprotin A treatment causes over a 30% rise in MK association with vasculature and a 20% increase in circulating platelets 24 hours later, with no change in MK number. Elevation of vascular SDF-1 by intravenous (IV) administration yields similar results. These data indicate that an endogenous SDF-1 gradient toward the vasculature contributes to homeostatic megakaryopoiesis and thrombopoiesis. At 2 days post-TBI, when endosteal SDF-1 message is increased, stabilization with Diprotin A results in a 40% decrease in MKs associated with vasculature and a small but significant decrease in platelets 24 hours later. Further supporting a role for altered SDF-1 gradients, elevating vascular levels with IV SDF-1 at 2 days causes the opposite effect of Diprotin A, with more MKs found in the vascular niche and a rise in peripheral platelet count. In contrast, at 3 days post-TBI, stabilization of endogenous SDF-1 with Diprotin A causes a further 25% increase in MKs in the vascular niche and a 10% rise in circulating platelets, consistent with the rapid loss of the endosteal SDF-1 gradient. Taken together, our data demonstrate that changes in microenvironmental SDF-1 regulate the spatial distribution of MKs in the post-TBI bone marrow. Importantly, the observed SDF-1 changes have functional consequences for platelet production, as the movement of MKs toward the endosteum decreases circulating platelets, while MK association with the vasculature increases circulating platelets. This knowledge will ultimately lead to improved therapeutic strategies to enhance platelet output in the setting of thrombocytopenia and highlights the need to carefully optimize the timing of therapeutic interventions. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
- Full Text
- View/download PDF
27. SDF-1 Acutely Promotes the Physical Association of Megakaryocytes with Vascular Endothelium in the Bone Marrow and Increases the Number of Circulating Platelets
- Author
-
Kathleen E. McGrath, Anne D. Koniski, James Palis, Jennifer L McLaughlin, and Lisa M. Niswander
- Subjects
medicine.medical_specialty ,Endothelium ,Chemistry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Endocrinology ,Megakaryocyte ,Precursor cell ,Internal medicine ,medicine ,Platelet ,Bone marrow ,Thrombopoiesis ,Thrombopoietin ,Megakaryopoiesis - Abstract
Abstract 2306 Thrombocytopenia complicates many diseases and can be a life-threatening consequence of genotoxic treatments including chemotherapy and radiation therapy. It is well established that thrombopoiesis occurs in the bone marrow where mature megakaryocyte (MK) precursor cells associate with sinusoidal endothelial cells and extrude pro-platelets into the vasculature. There has been much interest in elucidating mechanisms that control megakaryopoiesis and in utilizing these pathways to increase platelet output. The leading paradigm of megakaryopoiesis centers on the ability of cytokines, chiefly thrombopoietin (TPO), to promote MK progenitor proliferation and MK precursor maturation. More recently, attention has been focused on the ability of the bone marrow microenvironment to promote MK maturation and platelet formation. The chemokine stromal-derived factor-1 (SDF-1, also known as CXCL12), signaling through its receptor CXCR4, is implicated in the chemotaxis of MKs toward sinusoidal vessels, and in vivo evidence demonstrates that sustained plasma elevation of SDF-1 can increase platelet counts (Avecilla et al. Nature Medicine, 2004). To more specifically determine the short-term effects of SDF-1, we injected mice with a single 400ng intravenous dose of SDF-1 and enumerated the progenitor, precursor, and platelet compartments of the MK lineage. At 24 hours, SDF-1 induced a 30% increase in platelets compared to vehicle control (p0.7 and p>0.5). To quantitatively determine if SDF-1 regulates the physical interactions of MK precursors with sinusoidal endothelium, we developed a double immunohistochemistry assay using Gp1Bβ to distinguish MK precursors and MECA32 to identify vascular endothelial cells. In vehicle-treated mice, 39% of MKs in the marrow localized to the sinusoidal endothelium, and this increased to 53% 24 hours following SDF-1 treatment (p0.4 and p>0.7). As the platelet response following SDF-1 was less robust in the setting of TBI injury, we hypothesized that SDF-1-induced thrombopoiesis may improve if the number of MK precursors available to move to the vascular niche is increased. To test this, we administered TPO and SDF-1 at 2 hours and at 4 days, respectively, following TBI. TPO treatment alone resulted in 15% more MK precursors than irradiated vehicle controls at 5 days post-TBI (p0.2). In contrast, mice receiving both TPO and SDF-1 had over 20% more platelets than irradiated vehicle controls (p0.9). Correcting for differences in MK precursor numbers, mice receiving TPO and SDF-1 post-TBI had 1.8-fold more MKs in the vascular niche than irradiated vehicle controls (p Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
- Full Text
- View/download PDF
28. Abstract 3401: Genetic down-regulation of MET alters the metastatic phenotype of osteosarcoma cells
- Author
-
Su Young Kim, Lisa M. Niswander, James G. Christensen, Lillian M. Guenther, Chand Khanna, Lee J. Helman, Alessio Giubellino, Donald P. Bottaro, and Fabiola Cecchi
- Subjects
Cancer Research ,Gene knockdown ,biology ,business.industry ,medicine.disease ,Receptor tyrosine kinase ,Metastasis ,Small hairpin RNA ,Oncology ,Cell culture ,Immunology ,biology.protein ,Cancer research ,Medicine ,Osteosarcoma ,Immunohistochemistry ,business ,Tyrosine kinase - Abstract
The MET tyrosine kinase receptor is important during human development for normal cell growth and migration. Activating alterations of MET have also been identified in several carcinomas. Previous immunohistochemistry studies in our laboratory found high levels of MET in 85% of osteosarcoma tumor samples. In addition, pharmacologic modulation of MET resulted in a migratory decrease in some osteosarcoma cell lines. This led us to hypothesize that MET is necessary for osteosarcoma metastasis, and genetic down-regulation of MET expression would reduce the metastatic phenotype of osteosarcoma cells. We used lentiviral shRNA constructs to knockdown MET expression in several osteosarcoma cell lines. These included MNNG-HOS, a metastatic chemically transformed line that expresses constitutively activated MET, MG63.2, a highly metastatic variant of poorly metastatic MG63, AI and LR, which are two newly derived cell lines that have extremely high MET levels. We quantified levels of MET expression using a sensitive and quantitative electrochemiluminescence (ECL) assay and assessed in vitro parameters of metastasis including motility, migration, and invasion. MET expression was assessed by ECL in protein lysates from frozen osteosarcoma tumor samples and cell lines, and 15-20% of samples showed extremely high levels of total MET. AI, LR and MG63.2 had the highest levels of MET expression. MNNG-HOS has activation of MET due to translocation of the 3′end of MET with the 5′end of TPR. We utilized these lines for shRNA knock down of MET. ECL quantification of total MET levels revealed greater than 70% decrease in MET expression with shRNA knockdown in all four cell lines, compared with cells infected with scramble shRNA controls. As predicted for MNNG-HOS, shRNA constructs targeting the 3’ tyrosine kinase domain of MET resulted in marked decreased motility, compared to intermediate decreases using shRNA constructs targeting the 5’ region. Knock down of MET in MG63.2 resulted in inhibition of both motility and migration. AI and LR assays are in progress. Taken together, these data are the first evidence that genetic down-regulation of MET results in a decreased metastatic phenotype in osteosarcoma. This is a promising finding, as several small molecule tyrosine kinase inhibitors targeting MET are in development. We are currently testing our hypothesis in a mouse model to determine the ability of the above genetically knocked-down MET cell lines to decrease metastasis in vivo. Concurrently, in preparation for translational studies, we are also assessing MET specific tyrosine kinase inhibitors utilizing the parental cell lines described above. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3401.
- Published
- 2010
- Full Text
- View/download PDF
29. Mutation in Mouse Hei10, an E3 Ubiquitin Ligase, Disrupts Meiotic Crossing-over
- Author
-
Jeremy O Ward, Laura G Reinholdt, William W Motley, Lisa M Niswander, Dekker C Deacon, Laurie Beth Griffin, Kristofor Langlais, Vickie L Backus, Kerry Schimenti, Marilyn J O'Brien, John J Eppig, and John C. Schimenti
- Subjects
Cancer Research ,Genetics ,Molecular Biology ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics - Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.