Network


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

Hotspot


Dive into the research topics where Jane S. Reese is active.

Publication


Featured researches published by Jane S. Reese.


Journal of Clinical Investigation | 2003

In vivo selection of MGMT(P140K) lentivirus–transduced human NOD/SCID repopulating cells without pretransplant irradiation conditioning

Steven P. Zielske; Jane S. Reese; Karen Lingas; Jon Donze; Stanton L. Gerson

Infusion of transduced hematopoietic stem cells into nonmyeloablated hosts results in ineffective in vivo levels of transduced cells. To increase the proportion of transduced cells in vivo, selection based on P140K O6-methylguanine-DNA-methyltransferase (MGMT[P140K]) gene transduction and O6-benzylguanine/1,3-bis(2-chloroethyl)-1-nitrosourea (BG/BCNU) treatment has been devised. In this study, we transduced human NOD/SCID repopulating cells (SRCs) with MGMT(P140K) using a lentiviral vector and infused them into BG/BCNU-conditioned NOD/SCID mice before rounds of BG/BCNU treatment as a model for in vivo selection. Engraftment was not observed until the second round of BG/BCNU treatment, at which time human cells emerged to compose up to 20% of the bone marrow. Furthermore, 99% of human CFCs derived from NOD/SCID mice were positive for provirus as measured by PCR, compared with 35% before transplant and 11% in untreated irradiation-preconditioned mice, demonstrating selection. Bone marrow showed BG-resistant O6-alkylguanine-DNA-alkyltransferase (AGT) activity, and CFUs were stained intensely for AGT protein, indicating high transgene expression. Real-time PCR estimates of the number of proviral insertions in individual CFUs ranged from 3 to 22. Selection resulted in expansion of one or more SRC clones containing similar numbers of proviral copies per mouse. To our knowledge, these results provide the first evidence of potent in vivo selection of MGMT(P140K) lentivirus-transduced human SRCs following BG/BCNU treatment.


Human Gene Therapy | 1999

Delta MGMT-Transduced Bone Marrow Infusion Increases Tolerance to O6-Benzylguanine and 1,3-Bis(2-chloroethyl)1-nitrosourea and Allows Intensive Therapy of 1,3-Bis(2-chloroethyl)-1-nitrosourea-Resistant Human Colon Cancer Xenografts

Omer N. Koc; Jane S. Reese; Brian M. Davis; Lili Liu; Karen J. Majczenko; Stanton L. Gerson

O6-Benzylguanine (BG) is a potent inhibitor of the DNA repair protein 06-alkylguanine DNA alkyltransferase (AGT), and sensitizes tumors to BCNU in vitro and in xenografts. The combination of BG and BCNU is now undergoing phase I clinical testing. The maximally tolerated dose of BCNU given after BG is expected to be lower then the doses tolerated as a single agent owing to BG sensitization of hematopoietic progenitors. We have previously shown that retroviral expression of G156A mutant MGMT (deltaMGMT) in mouse and human marrow cells results in significant BG and BCNU resistance. In this study we evaluated the effect of deltaMGMT-transduced marrow infusion on the therapeutic index of multiple BG and BCNU treatments in tumor-bearing nude (nu/nu athymic) mice. Prior to subcutaneous implantation of BCNU-resistant SW480 human colon cancer cells, cohorts of mice were given intraperitoneal injections of nonablative doses of BG (30 mg/kg) and BCNU (10 mg/kg, one-half of the LD10) and then infused with 1-2 x 10(6) isogeneic deltaMGMT (n = 29 mice) or lacZ-transduced (n = 20 mice) marrow cells. The xenograft-bearing mice were treated with multiple cycles of BG (30 mg/kg) and BCNU (10-25 mg/kg). After three cycles, deltaMGMT mouse bone marrow was repopulated with CFU containing the provirus, and demonstrated a 2.7-fold increase in AGT activity and a 5.5-fold increase in BCNU IC90 compared with LacZ mice. After five cycles, the BCNU IC90 of CFU cells increased nine-fold over control cells, indicating selective enrichment of CFU precursor cells expressing high levels of deltaMGMT. Starting with the third cycle of therapy, tolerance to BG and BCNU was significantly improved in deltaMGMT mice compared with LacZ mice, as evidenced by preserved peripheral blood counts, bone marrow cellularity, and CFU content 1 and 2 weeks posttreatment and a significantly higher survival rate. Xenograft growth was significantly delayed in mice tolerating multiple cycles and higher dose intensity of BG and BCNU as compared with mice receiving less intensive therapy. We conclude that deltaMGMT-transduced marrow cells can improve the therapeutic index of BG and BCNU by selectively repopulating the marrow and providing significant marrow tolerance to this combination, allowing intensive therapy of a BCNU-resistant tumor.


Current Opinion in Hematology | 2010

Emerging therapeutic approaches for multipotent mesenchymal stromal cells

Paolo F. Caimi; Jane S. Reese; Zhenghong Lee; Hillard M. Lazarus

Purpose of reviewMultipotent mesenchymal stromal cells (MSCs) are rare cells resident in bone marrow and other organs capable of differentiating into mesodermal lineage tissues. MSCs possess immunomodulatory properties and have extensive capacity for ex-vivo expansion. Early clinical studies demonstrated safety and feasibility of infusing autologous MSCs and suggested a role in enhancing engraftment after hematopoietic cell transplant (HCT). Subsequent pilot studies using allogeneic MSCs showed safety but presented contradictory results regarding efficacy in treating graft-versus-host disease (GVHD). Recent findingsLarger, phase II allogeneic MSC infusion studies, including cells obtained from haploidentical and third-party donors, showed efficacy in GVHD treatment; however, recent randomized, placebo-controlled studies failed to corroborate these results. New investigations include MSC infusions in umbilical cord blood transplantation, MSC therapy for tissue regeneration/repair, harvest and use of MSCs from adipose tissue and cell-tracking/imaging studies using radionuclides, gene and fluorescent dye-labeled MSCs. SummaryMSCs remain the subject of intense investigation in HCT because of their differentiation potential and immunomodulatory properties. Whereas infusions of autologous, allogeneic and third-party donor MSCs are well tolerated, further research is needed to clarify the optimal methods for harvesting and expansion, optimal timing of administration and efficacy in the setting of HCT.


Journal of Hematotherapy & Stem Cell Research | 1999

Human Mesenchymal Stem Cells Provide Stromal Support for Efficient CD34+ Transduction

Jane S. Reese; Omer N. Koc; Stanton L. Gerson

Human mesenchymal stem cells (hMSC)-nonhematopoietic cells within the bone marrow microenvironment that can be culture expanded to a uniform population of fibroblastic cells-have been shown to support long-term hematopoiesis of CD34+ cells. Because direct contact between stromal elements and CD34+ cells enhances long-term engraftment, we postulated that hMSC would be a good alternative to the more heterogeneous stroma currently used in gene transfer studies. We used hMSC to support retroviral gene transfer of the G156A MGMT (deltaMGMT) gene encoding an alkyltransferase (AGT), which confers drug resistance to a combination of O6-benzylguanine (BG) plus the alkylating agents BCNU and temozolomide (TMZ) in human hematopoietic progenitors. In the presence of IL-3, IL-6, SCF, or leukemia inhibitory factor (LIF) and Flt-3 ligand, hMSC facilitated expansion and retroviral transduction of human peripheral blood-mobilized CD34+ cells. Furthermore, the transduced cells expressed AGT in 29% of hematopoietic cells and were 5-fold more resistant to BCNU and TMZ than were untransduced cells. Unirradiated hMSC present as support cells were simultaneously transduced and expressed AGT in 26% of the cells. Thus, the homogeneous nature of hMSC, and their ability to support gene transfer and be transduced themselves suggest they may be useful in clinical gene transfer protocols and have broad therapeutic applications.


Bone Marrow Transplantation | 2003

Hematopoietic stem cell gene therapy: progress toward therapeutic targets.

J L Vollweiler; S P Zielske; Jane S. Reese; Stanton L. Gerson

Summary:The concept of hematopoietic stem cell gene therapy is as exciting as that of stem cell transplantation itself. The past 20 years of research have led to improved techniques for transferring and expressing genes in hematopoietic stem cells and preclinical models now routinely indicate the ease with which new genes can be expressed in repopulating stem cells of multiple species. Both modified murine oncoretroviruses and lentiviruses transmit genes into the genome of hematopoietic stem cells and allow expression in the host following transplantation. Using oncoretroviruses, therapeutic genes for severe combined immunodeficiency, common variable gamma chain immunodeficiency, chronic granulomatous disease, Hurlers and Gauchers Disease have all been used clinically with only modest success except for the patients with immunodeficiency in whom a partial T-cell chimerism has been dramatic. Since stem cell selection in vivo appears important to the therapeutic success of gene transfer, drug resistance selection, most recently using the MGMT gene, has been developed and appears to be safe. Future trials combining a drug resistance and therapeutic gene are planned, as are trials using safety-modified lentiviruses. The therapeutic potential of hematopoietic stem cell gene therapy, particularly given recent advances in stem cell plasticity, remains an exceptionally exciting area of clinical research.


Molecular Therapy | 2003

Myeloablation is not required to select and maintain expression of the drug-resistance gene, mutant MGMT, in primary and secondary recipients.

Janice E. Bowman; Jane S. Reese; Karen Lingas; Stanton L. Gerson

Gene transduction of hematopoietic progenitors capable of reconstituting both primary and secondary recipients is an important milestone in preclinical development of gene therapy. Myeloablation conditioning prior to infusion of transduced stem cells causes significant host morbidity. In contrast, drug-resistance gene transfer utilizes judicious in vivo selection of transduced stem cells over time, reaching only the level of transduction and expression required. The O(6)-benzylguanine (BG)-resistant mutant O(6)-methylguanine-DNA methyltransferase (MGMT) gene is a potent selection gene for transduced cells. Using two different mutant MGMTs, G156A and P140K, that vary in BG resistance by a factor of 1:20, we asked whether long-term repopulating and secondary mouse-repopulating cells could be transduced, transplanted, and selected for in the nonmyeloablated recipient and whether the mutant MGMT would continue to be expressed in secondary recipient repopulating cells. We found that under stringent drug-selection competition, cells expressing the more BG-resistant variant, P140K-MGMT, were enriched over G156A-MGMT-expressing progenitors. In addition, the MFG retroviral vector transmitted the mutant MGMT gene to long-term repopulating cells that, after selective enrichment in the nonmyeloablated primary recipient, repopulated secondary mice and continued to express the transgene. Thus, MFG mutant MGMT vectors transduce repopulating hematopoietic stem cells that may be used both for chemotherapeutic drug resistance and to enrich for second therapeutic genes.


Journal of Hematotherapy & Stem Cell Research | 2001

MGMT Expression in Murine Bone Marrow Is a Major Determinant of Animal Survival After Alkylating Agent Exposure

Jane S. Reese; Xuisheng Qin; Christopher B. Ballas; Mutsuo Sekiguchi; Stanton L. Gerson

Myelosuppression is commonly observed after alkylating agent chemotherapy due to low levels of O(6)-alkylguanine DNA alkyltransferase protein (AGT) in hematopoietic progenitors. Mice that lack AGT in all organs, O(6)-methylguanine-DNA methyltransferase gene knockout (MGMT(-/-)) mice are extremely hypersensitive to the methylating agent N-methyl-N-nitrosourea (MNU) and exhibit a 10-fold reduction in the LD(90). To determine whether bone marrow damage was the cause of the increased lethality, we transplanted 1 x 10(6) wild-type marrow into MGMT(-/-) mice and MGMT(-/-) marrow into wild-type mice and observed survival after MNU. Lethally irradiated MGMT(-/-) mice given > or = 25 mg/kg MNU 3 weeks after transplant of wild-type cells survived > 30 days (n = 11), whereas this dose was lethal to control MGMT(-/-) mice 9-12 days post treatment (n = 5). Conversely, lethally irradiated wild-type mice transplanted with MGMT(-/-) cells died after only 20-60 mg/kg MNU within 8-12 days (n = 6). No significant toxicities were found in other organs. Additionally, in an in vivo post transplant competition model, wild-type long-term repopulating cells had a > 200-fold competitive survival advantage over MGMT(-/-) cells, and after MNU treatment completely repopulated the mouse when transplanted at only one-tenth the cell number. We also observed a strong selection for transplanted marrow-derived wild-type stromal elements in the MGMT(-/-) background after drug treatment. These data indicate that alkylating agent hypersensitivity of MGMT(-/-) mice results from hematopoietic damage at the stem level. Thus, DNA repair involving AGT in hematopoietic cells is required for normal host survival following exposure to methylating and chloroethylating agents.


Cancer Gene Therapy | 1999

Human long-term culture initiating cells are sensitive to benzylguanine and 1,3-bis(2-chloroethyl)-1-nitrosourea and protected after mutant (G156A) methylguanine methyltransferase gene transfer.

Omer N. Koc; Jane S. Reese; E. Szekely; Stanton L. Gerson

Human hematopoietic progenitors express low levels of O6-alkylguanine-DNA alkyltransferase and are sensitive to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), particularly following O6-benzylguanine (BG)-mediated O6-alkylguanine-DNA alkyltransferase inhibition. Expression of the BG-resistant mutant (G156A) methylguanine methyltransferase (ΔMGMT) gene in hematopoietic cells confers resistance to BG and BCNU. Because BCNU targets both early and late human hematopoietic cells and results in prolonged and cumulative myelosuppression, we attempted to protect early hematopoietic progenitors (long-term culture initiating cells (LTC-ICs)) by retroviral-mediated transfer of the ΔMGMT gene. A total of 33–56% of LTC-ICs were transduced with MFG-ΔMGMT retrovirus as determined by evidence of provirus in secondary colony-forming units at 5 weeks of culture under conditions optimal for the survival and proliferation of early hematopoietic progenitors. The addition of flt-3 ligand to cultures increased the transduction rate of LTC-ICs. Furthermore, 17.8 ± 8.1% of ΔMGMT-transduced LTC-ICs survived doses of BG and BCNU; these doses allowed the survival of only 0–1% of untransduced LTC-ICs. This finding compares favorably with the 8–12% of CD34+ cell-derived colony-forming units that we previously showed became resistant to BG and BCNU after ΔMGMT gene transfer. Thus, ΔMGMT transduction of human early hematopoietic progenitor LTC-ICs confers resistance to BG and BCNU and may allow transduced LTC-ICs selective survival and enrichment over untransduced cells in patients undergoing BG and BCNU chemotherapy.


Stem Cells Translational Medicine | 2016

Human Mesenchymal Stem Cells Impact Th17 and Th1 Responses Through a Prostaglandin E2 and Myeloid-Dependent Mechanism

Ayal Rozenberg; Ayman Rezk; Marie Noëlle Boivin; Peter J. Darlington; Mukanthu Nyirenda; Rui Li; Farzaneh Jalili; Raz Winer; Elinor A. Artsy; Antonio Uccelli; Jane S. Reese; Sarah M. Planchon; Jeffrey Cohen; Amit Bar-Or

Human mesenchymal stem cells (hMSCs) are being increasingly pursued as potential therapies for immune‐mediated conditions, including multiple sclerosis. Although they can suppress human Th1 responses, they reportedly can reciprocally enhance human Th17 responses. Here, we investigated the mechanisms underlying the capacity of hMSCs to modulate human Th1 and Th17 responses. Human adult bone marrow‐derived MSCs were isolated, and their purity and differentiation capacity were confirmed. Human venous peripheral blood mononuclear cells (PBMC) were activated, alone, together with hMSC, or in the presence of hMSC‐derived supernatants (sups). Cytokine expression by CD4+ T‐cell subsets (intracellular staining by fluorescence‐activated cell sorting) and secreted cytokines (enzyme‐linked immunosorbent assay) were then quantified. The contribution of prostaglandin E2 (PGE2) as well as of myeloid cells to the hMSC‐mediated regulation of T‐cell responses was investigated by selective depletion of PGE2 from the hMSC sups (anti‐PGE2 beads) and by the selective removal of CD14+ cells from the PBMC (magnetic‐activated cell sorting separation). Human MSC‐secreted products could reciprocally induce interleukin‐17 expression while decreasing interferon‐γ expression by human CD4+ T cells, both in coculture and through soluble products. Pre‐exposure of hMSCs to IL‐1β accentuated their capacity to reciprocally regulate Th1 and Th17 responses. Human MSCs secreted high levels of PGE2, which correlated with their capacity to regulate the T‐cell responses. Selective removal of PGE2 from the hMSC supernatants abrogated the impact of hMSC on the T cells. Selective removal of CD14+ cells from the PBMCs also limited the capacity of hMSC‐secreted PGE2 to affect T‐cell responses. Our discovery of a novel PGE2‐dependent and myeloid cell‐mediated mechanism by which human MSCs can reciprocally induce human Th17 while suppressing Th1 responses has implications for the use of, as well as monitoring of, MSCs as a potential therapeutic for patients with multiple sclerosis and other immune‐mediated diseases.


Stem Cells International | 2016

Antimicrobial Properties of Mesenchymal Stem Cells: Therapeutic Potential for Cystic Fibrosis Infection, and Treatment

Morgan T. Sutton; David Fletcher; Santosh K. Ghosh; Aaron Weinberg; Rolf van Heeckeren; Sukhmani Kaur; Zhina Sadeghi; Adonis Hijaz; Jane S. Reese; Hillard M. Lazarus; Donald P. Lennon; Arnold I. Caplan; Tracey L. Bonfield

Cystic fibrosis (CF) is a genetic disease in which the battle between pulmonary infection and inflammation becomes the major cause of morbidity and mortality. We have previously shown that human MSCs (hMSCs) decrease inflammation and infection in the in vivo murine model of CF. The studies in this paper focus on the specificity of the hMSC antimicrobial effectiveness using Pseudomonas aeruginosa (gram negative bacteria) and Staphylococcus aureus (gram positive bacteria). Our studies show that hMSCs secrete bioactive molecules which are antimicrobial in vitro against Pseudomonas aeruginosa, Staphylococcus aureus, and Streptococcus pneumonia, impacting the rate of bacterial growth and transition into colony forming units regardless of the pathogen. Further, we show that the hMSCs have the capacity to enhance antibiotic sensitivity, improving the capacity to kill bacteria. We present data which suggests that the antimicrobial effectiveness is associated with the capacity to slow bacterial growth and the ability of the hMSCs to secrete the antimicrobial peptide LL-37. Lastly, our studies demonstrate that the tissue origin of the hMSCs (bone marrow or adipose tissue derived), the presence of functional cystic fibrosis transmembrane conductance regulator (CFTR: human, Cftr: mouse) activity, and response to effector cytokines can impact both hMSC phenotype and antimicrobial potency and efficacy. These studies demonstrate, the unique capacity of the hMSCs to manage different pathogens and the significance of their phenotype in both the antimicrobial and antibiotic enhancing activities.

Collaboration


Dive into the Jane S. Reese's collaboration.

Top Co-Authors

Avatar

Stanton L. Gerson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Karen Lingas

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Omer N. Koc

University Hospitals of Cleveland

View shared research outputs
Top Co-Authors

Avatar

Brian M. Davis

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Hillard M. Lazarus

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Lili Liu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Boro Dropulic

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Justin C. Roth

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Pingfu Fu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge