Network


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

Hotspot


Dive into the research topics where Susan K. Dunn is active.

Publication


Featured researches published by Susan K. Dunn.


Blood | 2012

Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors

Daniel González-Nieto; Lina Li; Anja Köhler; Gabriel Ghiaur; Eri Taniguchi Ishikawa; Amitava Sengupta; Malav Madhu; Jorden Arnett; Rebecca A. Santho; Susan K. Dunn; Glenn I. Fishman; David E. Gutstein; Roberto Civitelli; Luis C. Barrio; Matthias Gunzer; Jose A. Cancelas

Connexin-43 (Cx43), a gap junction protein involved in control of cell proliferation, differentiation and migration, has been suggested to have a role in hematopoiesis. Cx43 is highly expressed in osteoblasts and osteogenic progenitors (OB/P). To elucidate the biologic function of Cx43 in the hematopoietic microenvironment (HM) and its influence in hematopoietic stem cell (HSC) activity, we studied the hematopoietic function in an in vivo model of constitutive deficiency of Cx43 in OB/P. The deficiency of Cx43 in OB/P cells does not impair the steady state hematopoiesis, but disrupts the directional trafficking of HSC/progenitors (Ps) between the bone marrow (BM) and peripheral blood (PB). OB/P Cx43 is a crucial positive regulator of transstromal migration and homing of both HSCs and progenitors in an irradiated microenvironment. However, OB/P Cx43 deficiency in nonmyeloablated animals does not result in a homing defect but induces increased endosteal lodging and decreased mobilization of HSC/Ps associated with proliferation and expansion of Cxcl12-secreting mesenchymal/osteolineage cells in the BM HM in vivo. Cx43 controls the cellular content of the BM osteogenic microenvironment and is required for homing of HSC/Ps in myeloablated animals.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells

Eri Taniguchi Ishikawa; Daniel González-Nieto; Gabriel Ghiaur; Susan K. Dunn; Ashley M. Ficker; Bhuvana Murali; Malav Madhu; David E. Gutstein; Glenn I. Fishman; Luis C. Barrio; Jose A. Cancelas

Hematopoietic stem cell (HSC) aging has become a concern in chemotherapy of older patients. Humoral and paracrine signals from the bone marrow (BM) hematopoietic microenvironment (HM) control HSC activity during regenerative hematopoiesis. Connexin-43 (Cx43), a connexin constituent of gap junctions (GJs) is expressed in HSCs, down-regulated during differentiation, and postulated to be a self-renewal gene. Our studies, however, reveal that hematopoietic-specific Cx43 deficiency does not result in significant long-term competitive repopulation deficiency. Instead, hematopoietic Cx43 (H-Cx43) deficiency delays hematopoietic recovery after myeloablation with 5-fluorouracil (5-FU). 5-FU-treated H-Cx43-deficient HSC and progenitors (HSC/P) cells display decreased survival and fail to enter the cell cycle to proliferate. Cell cycle quiescence is associated with down-regulation of cyclin D1, up-regulation of the cyclin-dependent kinase inhibitors, p21cip1. and p16INK4a, and Forkhead transcriptional factor 1 (Foxo1), and activation of p38 mitogen-activated protein kinase (MAPK), indicating that H-Cx43-deficient HSCs are prone to senescence. The mechanism of increased senescence in H-Cx43-deficient HSC/P cells depends on their inability to transfer reactive oxygen species (ROS) to the HM, leading to accumulation of ROS within HSCs. In vivo antioxidant administration prevents the defective hematopoietic regeneration, as well as exogenous expression of Cx43 in HSC/P cells. Furthermore, ROS transfer from HSC/P cells to BM stromal cells is also rescued by reexpression of Cx43 in HSC/P. Finally, the deficiency of Cx43 in the HM phenocopies the hematopoietic defect in vivo. These results indicate that Cx43 exerts a protective role and regulates the HSC/P ROS content through ROS transfer to the HM, resulting in HSC protection during stress hematopoietic regeneration.


Blood | 2012

Vav3 collaborates with p190-BCR-ABL in lymphoid progenitor leukemogenesis, proliferation, and survival.

Kyung Hee Chang; Abel Sanchez-Aguilera; Shuhong Shen; Amitava Sengupta; Malav Madhu; Ashley M. Ficker; Susan K. Dunn; Ashley M. Kuenzi; Jorden Arnett; Rebecca A. Santho; Xabier Agirre; John P. Perentesis; Michael W. Deininger; Yi Zheng; Xosé R. Bustelo; David A. Williams; Jose A. Cancelas

Despite the introduction of tyrosine kinase inhibitor therapy, the prognosis for p190-BCR-ABL(+) acute lymphoblastic leukemia remains poor. In the present study, we present the cellular and molecular roles of the Rho GTPase guanine nucleotide exchange factor Vav in lymphoid leukemogenesis and explore the roles of Vav proteins in BCR-ABL-dependent signaling. We show that genetic deficiency of the guanine nucleotide exchange factor Vav3 delays leukemogenesis by p190-BCR-ABL and phenocopies the effect of Rac2 deficiency, a downstream effector of Vav3. Compensatory up-regulation of expression and activation of Vav3 in Vav1/Vav2-deficient B-cell progenitors increases the transformation ability of p190-BCR-ABL. Vav3 deficiency induces apoptosis of murine and human leukemic lymphoid progenitors, decreases the activation of Rho GTPase family members and p21-activated kinase, and is associated with increased Bad phosphorylation and up-regulation of Bax, Bak, and Bik. Finally, Vav3 activation only partly depends on ABL TK activity, and Vav3 deficiency collaborates with tyrosine kinase inhibitors to inhibit CrkL activation and impair leukemogenesis in vitro and in vivo. We conclude that Vav3 represents a novel specific molecular leukemic effector for multitarget therapy in p190-BCR-ABL-expressing acute lymphoblastic leukemia.


Transfusion | 2013

A randomized controlled trial evaluating recovery and survival of 6% dimethyl sulfoxide–frozen autologous platelets in healthy volunteers

Larry J. Dumont; Jose A. Cancelas; Deborah F. Dumont; Alan Siegel; Zbigniew M. Szczepiorkowski; Neeta Rugg; P. Gayle Pratt; D. Nicole Worsham; Elizabeth L. Hartman; Susan K. Dunn; Margaret O'Leary; Janet H. Ransom; Rodney A. Michael; Victor W. Macdonald

BACKGROUND: Availability of platelets (PLTs) is severely limited by shelf life in some settings. Our objective was to determine and compare to Food and Drug Administration (FDA) criteria the PLT recovery and survival of autologous PLTs cryopreserved at −65°C or less in 6% dimethyl sulfoxide (DMSO) reconstituted with a no‐wash method (cryopreserved PLTs [CPPs]) compared to autologous fresh PLTs.


Blood | 2010

Rac2 GTPase deficiency depletes BCR-ABL+ leukemic stem cells and progenitors in vivo.

Amitava Sengupta; Jorden Arnett; Susan K. Dunn; David A. Williams; Jose A. Cancelas

Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disease (MPD) initiated by p210-BCR-ABL-mediated transformation of hematopoietic stem cells (HSCs). Inhibition of the ABL kinase alone is not sufficient to eradicate leukemic stem cells (LSCs). We have previously shown that the deficiency of Rac2 GTPase signaling, but not Rac1, in p210-BCR-ABL-transduced hematopoietic cells prolonged survival of mice with MPD. Here we demonstrate that absence of Rac2 GTPase prolongs survival of HSC-initiated, inducible Scl/p210-BCR-ABL (Scl/p210) binary transgenic mice, it induces apoptosis, and, unlike in normal HSC and progenitor (HSC/P), impairs LSC and progenitor (LSC/P) proliferation in vivo. As a result, Rac2 deficiency causes functional exhaustion of the LSC pool in vivo. This defect is not due to impaired interaction with the hematopoietic microenvironment as reflected by its unaltered adhesion, migration, and homing to recipient organs. In summary, Rac2 deficiency exhausts the LSC pool in vivo through impairment of oncogene-induced proliferation and survival signals.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Atypical protein kinase C (aPKCζ and aPKCλ) is dispensable for mammalian hematopoietic stem cell activity and blood formation

Amitava Sengupta; Angeles Duran; Eri Ishikawa; Maria Carolina Florian; Susan K. Dunn; Ashley M. Ficker; Michael Leitges; Hartmut Geiger; Maria T. Diaz-Meco; Jorge Moscat; Jose A. Cancelas

The stem-cell pool is considered to be maintained by a balance between symmetric and asymmetric division of stem cells. The cell polarity model proposes that the facultative use of symmetric and asymmetric cell division is orchestrated by a polarity complex consisting of partitioning-defective proteins Par3 and Par6, and atypical protein kinase C (aPKCζ and aPKCλ), which regulates planar symmetry of dividing stem cells with respect to the signaling microenvironment. However, the role of the polarity complex is unexplored in mammalian adult stem-cell functions. Here we report that, in contrast to accepted paradigms, polarization and activity of adult hematopoietic stem cell (HSC) do not depend on either aPKCζ or aPKCλ or both in vivo. Mice, having constitutive and hematopoietic-specific (Vav1-Cre) deletion of aPKCζ and aPKCλ, respectively, have normal hematopoiesis, including normal HSC self-renewal, engraftment, differentiation, and interaction with the bone marrow microenvironment. Furthermore, inducible complete deletion of aPKCλ (Mx1-Cre) in aPKCζ−/− HSC does not affect HSC polarization, self-renewal, engraftment, or lineage repopulation. In addition, aPKCζ- and aPKCλ-deficient HSCs elicited a normal pattern of hematopoietic recovery secondary to myeloablative stress. Taken together, the expression of aPKCζ, aPKCλ, or both are dispensable for primitive and adult HSC fate determination in steady-state and stress hematopoiesis, contrary to the hypothesis of a unique, evolutionary conserved aPKCζ/λ-directed cell polarity signaling mechanism in mammalian HSC fate determination.


Transfusion | 2011

In vivo viability of stored red blood cells derived from riboflavin plus ultraviolet light-treated whole blood

Jose A. Cancelas; Neeta Rugg; Dana Fletcher; P. Gayle Pratt; D. Nicole Worsham; Susan K. Dunn; Susanne Marschner; Heather L. Reddy; Raymond P. Goodrich

BACKGROUND: A novel system using ultraviolet (UV) light and riboflavin (Mirasol System, CaridianBCT Biotechnologies) to fragment nucleic acids has been developed to treat whole blood (WB), aiming at the reduction of potential pathogen load and white blood cell inactivation. We evaluated stored red blood cell (RBC) metabolic status and viability, in vitro and in vivo, of riboflavin/UV light–treated WB (IMPROVE study).


Nature Communications | 2013

Klf5 controls bone marrow homing of stem cells and progenitors through Rab5-mediated β1/β2-integrin trafficking

E. Taniguchi Ishikawa; K. H. Chang; Ramesh Nayak; H. A. Olsson; Ashley M. Ficker; Susan K. Dunn; Malav Madhu; Amitava Sengupta; Jeffrey A. Whitsett; H. L. Grimes; Jose A. Cancelas

Kruppel-like factor 5 (Klf5) regulates pluripotent stem cell self-renewal but its role in somatic stem cells is unknown. Here we show that Klf5 deficient haematopoietic stem cells and progenitors (HSC/P) fail to engraft after transplantation. This HSC/P defect is associated with impaired bone marrow homing and lodging and decreased retention in bone marrow, and with decreased adhesion to fibronectin and expression of membrane-bound β1/β2-integrins. In vivo inducible gain-of-function of Klf5 in HSCs increases HSC/P adhesion. The expression of Rab5 family members, mediators of β1/β2-integrin recycling in the early endosome, is decreased in Klf5Δ/Δ HSC/Ps. Klf5 binds directly to the promoter of Rab5a/b and overexpression of Rab5b rescues the expression of activated β1/β2-integrins, adhesion and bone marrow homing of Klf5Δ/Δ HSC/Ps. Altogether, these data indicate that Klf5 is indispensable for adhesion, homing, lodging and retention of HSC/Ps in the bone marrow through Rab5-dependent post-translational regulation of β1/β2 integrins.


Blood | 2012

Bmi1 reprograms CML B-lymphoid progenitors to become B-ALL–initiating cells

Amitava Sengupta; Ashley M. Ficker; Susan K. Dunn; Malav Madhu; Jose A. Cancelas

The characterization and targeting of Philadelphia chromosome positive (Ph(+)) acute lymphoblastic leukemia (ALL)-initiating cells remains unresolved. Expression of the polycomb protein Bmi1 is up-regulated in patients with advanced stages of chronic myelogenous leukemia (CML). We report that Bmi1 transforms and reprograms CML B-lymphoid progenitors into stem cell leukemia (Scl) promoter-driven, self-renewing, leukemia-initiating cells to result in B-lymphoid leukemia (B-ALL) in vivo. In vitro, highly proliferating and serially replatable myeloid and lymphoid colony-forming cultures could be established from BCR-ABL and Bmi1 coexpressing progenitors. However, unlike in vivo expanded CML B-lymphoid progenitors, hematopoietic stem cells, or multipotent progenitors, coexpressing BCR-ABL and Bmi1 did not initiate or propagate leukemia in a limiting dilution assay. Inducible genetic attenuation of BCR-ABL reversed Bmi1-driven B-ALL development, which was accompanied by induction of apoptosis of leukemic B-lymphoid progenitors and by long-term animal survival, suggesting that BCR-ABL is required to maintain B-ALL and that BCR-ABL and Bmi1 cooperate toward blast transformation in vivo. Our data indicate that BCR-ABL targeting itself is required to eradicate Ph(+)/Bmi1(+) B-ALL-initiating cells and confirm their addiction to BCR-ABL signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells (Proceedings of the National Academy of Sciences (2012) 109, (9071-9076) DOI: 10.1073/pnas. 1120358109)

Eri Ishikawa; Daniel González-Nieto; Gabriel Ghiaur; Susan K. Dunn; Ashley M. Ficker; Bhuvana Murali; Malav Madhu; David E. Gutstein; Glenn I. Fishman; Luis C. Barrio; Jose A. Cancelas

Collaboration


Dive into the Susan K. Dunn's collaboration.

Top Co-Authors

Avatar

Jose A. Cancelas

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amitava Sengupta

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ashley M. Ficker

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jorden Arnett

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Malav Madhu

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Daniel González-Nieto

Technical University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Gabriel Ghiaur

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luis C. Barrio

Autonomous University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge