GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2419-2419
    Abstract: Hematopoietic stem and progenitor cell (HSPC) migration, marrow homing and engraftment are key physiological processes regulating hematopoiesis post transplantation. These processes are the result of the orchestrated actions of multiple extracellular stimuli, which regulate actin remodeling, cell polarity, chemotaxis and cell-cell interactions. In HSPC, the Rho GTPases Rac and CDC42 act as molecular switches that integrate extracellular stimuli in a spatially regulated manner to control cell migration and mediate homing to marrow and mobilization as well as cell survival/ proliferation pathways to mediate engraftment(Gu et al., Science 2003; Cancelas et al., Nature Medicine 2005; Wang et al., Blood 2006). Using an inhibitory peptide against Group A p21 activated kinases (Pak1-3), key effectors of Rac/ CDC42 and individual Pak1 & 2 genetic knock-out mice, we recently demonstrated that Pak kinases, specifically Pak2, are important for HSPC homing and engraftment (Dorrance et al., Blood 2013). Pak2 is a multi-domain protein that contains a C-terminal kinase domain and multiple N-terminal protein-interaction domains. Among these is a non-classical SH3-binding site for the guanine-nucleotide-exchange factor β-PIX, which was shown to be critical for both activation of Rac1 and its localization to and induction of membrane ruffles (Klooster et al., Journal of Cell Biology 2006). In this study we further explored the role of these domains of Pak2 in key HSPC functions, including homing to bone marrow in vivo. We employed a multi-cistronic retrovirus vector that simultaneously deleted floxed endogenous Pak2 gene sequences and rescued with either wild type (WT), a kinase dead (KD) mutant (K278A, defective in auto/ trans phosphorylation) or a Δβ-PIX mutant, (P185/R186A, that cannot bind to β-PIX). As previously demonstrated deletion of Pak2 (Pak2 Δ/Δ) was associated with abnormal SDF-1 stimulated cell protrusions containing F-actin (as demonstrated by confocal and electron microscopy) and these HSPC displayed decreased directional migration (Euclidean distance in Pak2Δ/Δ vs. Pak2WT/WT: 39.6µm ±9.6 vs. 96.6µm ±21.6; P 〈 0.05). This phenotype of abnormal cell protrusions and decreased directional migration was rescued by expressing Pak2-WT (Pak2WT/WT vs. Pak2-WT: 96.6µm ±21.6 vs. 74.0µm ±18.7; P: not significant) but not by expressing Pak2-KD (Pak2WT/WT vs. Pak2-KD: 96.6µm ±21.6 vs. 33.6µm ±6.3; P 〈 0.05) demonstrating the requirement of Pak2 kinase activity in SDF1-induced cell polarization and directed cell migration. Interestingly, we found abnormal F-actin clustering associated with defective polarization (by confocal microscopy) and decreased velocity of cell migration in time-lapsed video microscopy when Pak2-deletion was rescued with Pak2-Δβ-PIX (velocity of migration Pak2WT/WT vs. Pak2-Δβ-PIX, 0.32µm/minute ±0.02 vs. 0.13µm/minute ±0.02; P 〈 0.001), indicating the requirement of β-PIX exchange factor interaction with Pak2 in directed migration. To test whether these in vitro phenotypes were associated with changes in homing efficiency to bone marrow, we performed in vivo homing assays of rescued HSPC. Transduced, GFP-sorted Lin-Sca1+Kit+ cells of each genotype were injected into lethally-irradiated C57BL/6 recipient mice (N= 12-29 /genotype). Twelve hours post-transplantation the number of EGFP+ cells in the bone marrow was determined and percent homing is calculated. Compared to Pak2 WT/WT, Pak2Δ/Δ HSPC displayed reduced homing (99.26%± 4.9 vs. 53.4% ± 4.2; P 〈 0.0001). The homing defect was rescued by Pak2-WT (Pak2WT/WT vs. Pak2-WT rescue: 99.26%± 4.9 vs. 86% ± 8.5; P: not significant). However neither Pak2-KD nor Pak2-Δβ-PIX rescued in vivo homing: 99.26% ±4.9 vs. 38.9% ±3.7 vs. 33.0%± 6.0; P 〈 0.0001 each mutant vs.Pak2WT/WT) proving the necessity of kinase activity and interaction with β-PIX for bone marrow homing. Taken together we show that both Pak2-kinase activity and its interaction with β-PIX exchange factor are required for coordinated HSPC F-actin formation and cell polarization, directed cell migration in vitro and homing to bone marrow in vivo. These data directly link the in vitro effects of Pak2 kinase with in vivo bonemarrow homing. Note All p values are calculated by Mann Whitney test. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Clinical Cancer Research Vol. 21, No. 17_Supplement ( 2015-09-01), p. A40-A40
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 17_Supplement ( 2015-09-01), p. A40-A40
    Abstract: Megakaryocytes mature and differentiate into polyploid cells in the bone marrow, and migrate from the osteoblastic niche to the vascular niche by means of an SDF-1α gradient, culminating in the release of platelets into circulation. Failure to do so results in myeloproliferative disorders, which can manifest as thrombocytopenia. Many chemotherapeutic agents are known to induce thrombocytopenia and other platelet disorders. Regulation of megakaryocyte differentiation and polyploidization is controlled by MAPK associated proteins, and cytoskeleton regulating proteins. p21-activated kinase 2 (Pak2) is known to regulate these two pathways, but nothing is currently known about the role of Pak2 in megakaryocyte biology. In our study, we sought to demonstrate that Pak2 is essential for proper megakaryocyte differentiation, maturation and polyploidization, in vivo and in vitro. Inducible deletion of Pak2fl/fl results in 100% lethality within 20 days of cre induction, specifically in hematopoietic tissues (Mx1-cre). Peripheral blood analysis demonstrates thrombocytopenia (reduced platelet counts), along with increased neutrophil count (neutrophilia) and decreased lymphocyte count. Bone marrow histology and analysis by flow cytometry demonstrates altered megakaryocyte differentiation, observed by elevated megakaryocyte(Mk) precursors in the bone marrow (Lin-ckit+Sca1- CD150+CD41+), elevated mature bone marrow Mks (CD41+), increased number of polyploid cells( & gt;8N) and hyperplastic megakaryocytes. In vitro culture of bone marrow derived Pak2-null megakaryocytes demonstrates elevated polyploidization, altered β1-tubulin structure and reduced platelet formation. Additionally, cre activation in platelets and megakaryocytes using a PF4-cre driver (platelet factor 4) recapitulates the enhanced polyploidization phenotype observed in the Mx1-cre model. Ultimately, we demonstrate that Pak2 signals through LIMK, cofilin, and myosin light chain 2 (MLC2) to regulate megakaryocyte cytoskeleton during differentiation and polyploidization. In summary, Pak2 is an important regulator of megakaryocyte maturation and platelet formation in vivo via regulation of proteins involved in cytoskeletal dynamics. Citation Format: Rachelle E. Kosoff, Jonathan Chernoff. Megakaryocyte endomitosis requires Pak2 to regulate actin and microtubule networks. [abstract]. In: Proceedings of the AACR Special Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; Sep 20-23, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Clin Cancer Res 2015;21(17 Suppl):Abstract nr A40.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Molecular Cancer Research Vol. 12, No. 12_Supplement ( 2014-12-01), p. A15-A15
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 12, No. 12_Supplement ( 2014-12-01), p. A15-A15
    Abstract: Megakaryocytes (MKs) mature and differentiate into multi-nucleated cells in the bone marrow, and migrate from the osteoblastic niche to the vascular niche by means of an SDF-1a gradient, culminating in the release of platelets into circulation. Failure to do so results in myeloproliferative disorders, which can manifest with extremely reduced platelet counts. Regulation of this process is controlled by MAPK signaling, cytoskeletal and microtubule-associated proteins and GTPases. p21-activated kinase 2 (Pak2) regulates the cytoskeleton through the MAPK and RhoA signaling pathways, both required for MK differentiation and cytokinesis. In our study, we demonstrate that Pak2 is essential for proper MK differentiation and polyploidization, in vitro and in vivo. Pak2 deletion, upon cre induction specifically in hematopoietic tissues (Mx1-cre), reduces peripheral blood platelet counts (thrombocytopenia) by 50%. Bone marrow histology demonstrates hyperplastic MKs coinciding with reduced platelet counts. Analysis of bone marrow by flow cytometry demonstrates altered MK differentiation, observed by elevated MK stem cell precursors in the bone marrow (CD150+CD41+). Increased stem cell potential of MKs deficient for Pak2 results in increased colony forming units (CFU-Mks). Additionally, Pak2-null MKs have increased levels of polyploidization. Signaling to MLC2 and GEF-H1 are altered in Pak2-deficient MKs, both previously shown to regulate MK differentiation and polyploidization, respectively. In summary, Pak2 is a negative regulator of MK stem cell differentiation as well as polyploidization. Since Pak2 negatively regulates polyploidization, this leads to therapeutic uses for Pak family inhibitors in the treatment for acute megakaryoblastic leukemia, a fatal disorder which presents with an accumulation of immature MKs, due to a failure to differentiate into mature MKs. Citation Format: Rachelle E. Kosoff, Jonathan Chernoff. Megakaryocyte differentiation is regulated by p21-activated kinase 2. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr A15. doi: 10.1158/1557-3125.RASONC14-A15
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2098788-2
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: The EMBO Journal, Springer Science and Business Media LLC, Vol. 30, No. 15 ( 2011-7-1), p. 3160-3172
    Type of Medium: Online Resource
    ISSN: 0261-4189 , 1460-2075
    RVK:
    Language: Unknown
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 1467419-1
    detail.hit.zdb_id: 586044-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Wiley ; 2012
    In:  Journal of Cellular Physiology Vol. 227, No. 6 ( 2012-06), p. 2654-2659
    In: Journal of Cellular Physiology, Wiley, Vol. 227, No. 6 ( 2012-06), p. 2654-2659
    Abstract: Understanding the molecular and cellular processes underlying the development, maintenance, and progression of Barrett's esophagus (BE) presents an empirical challenge because there are no simple animal models and standard 2D cell culture can distort cellular processes. Here we describe a three‐dimensional (3D) cell culture system to study BE. BE cell lines (CP‐A, CP‐B, CP‐C, and CP‐D) and esophageal squamous keratinocytes (EPC2) were cultured on a matrix consisting of esophageal fibroblasts and collagen. Comparison of growth and cytokeratin expression in the presence of all‐ trans retinoic acid or hydrochloric acid was made by immunohistochemistry and Alcian Blue staining to determine which treatments produced a BE phenotype of columnar cytokeratin expression in 3D culture. All‐ trans retinoic acid differentially affected the growth of BE cell lines in 3D culture. Notably, the non‐dyplastic metaplasia‐derived cell line (CP‐A) expressed reduced squamous cytokeratins and enhanced columnar cytokeratins upon ATRA treatment. ATRA altered the EPC2 squamous cytokeratin profile towards a more columnar expression pattern. Cell lines derived from patients with high‐grade dysplasia already expressed columnar cytokeratins and therefore did not show a systematic shift toward a more columnar phenotype with ATRA treatment. ATRA treatment, however, did reduce the squamoid‐like multilayer stratification observed in all cell lines. As the first study to demonstrate long‐term 3D growth of BE cell lines, we have determined that BE cells can be cultured for at least 3 weeks on a fibroblast/collagen matrix and that the use of ATRA causes a general reduction in squamous‐like multilayered growth and an increase in columnar phenotype with the specific effects cell‐line dependent. J. Cell. Physiol. 227: 2654–2659, 2012. © 2011 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 0021-9541 , 1097-4652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 1478143-8
    detail.hit.zdb_id: 3116-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Elsevier BV ; 2013
    In:  Journal of Biological Chemistry Vol. 288, No. 2 ( 2013-01), p. 974-983
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 288, No. 2 ( 2013-01), p. 974-983
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 2997-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 917-917
    Abstract: Abstract 917 Rho GTPases, including Rac, integrate multiple extracellular signals and play important regulatory roles in HSC/P functions such as engraftment, retention, migration, adhesion, proliferation, and survival (Gu et al. Science, 2003). Our studies now focus on identifying potential Rac downstream effector proteins important for normal HSC/P function(s). The p21-activated kinases (Pak) are serine/threonine kinases that interact with and are major downstream targets of Rac and Cdc42. There are six human Paks (Pak1–6), which are grouped based on homology into Group A (Pak 1–3) and Group B (4–6) Paks. Paks regulate cytoskeletal organization including stress fiber dissolution, lamellipodia formation and focal adhesion disassembly and mediate activation of MAPK pathways. To identify the possible role(s) of Pak proteins in engraftment, freshly isolated LSK cells from WT (CD45.1+/CD45.2+) BM were transduced with retrovirus containing the Pak Inhibitory Domain (PID), which inhibits Group A Pak protein function or empty vector control (Mieg3); both constructs co-express GFP. 1.0×105 GFP+ LSK+ cells were then isolated and co-transplanted with 5.0×105 BoyJ (CD45.1+) whole bone marrow (WBM) into lethally irradiated C57Bl/6J (CD45.2+) recipients. Percent chimerism was measured at 3- to 24- weeks post BMT. PID transduced LSK+ cells were incapable of contributing to recipient hematopoietic reconstitution (Table 1). To explore the underlying mechanism of this engraftment failure we performed in vivo homing assays and found a 4- and 16- fold decrease, respectively, in BM homing of PID transduced LSK+ vs controls at 12 and 48 hours (p 〈 0.05, for both time points). Altered cell migration of LSK+ cells was confirmed by live imaging microscopy which showed a 4-fold decrease in overall cell displacement in SDF-1-stimulated directed migration in the PID-expressing LSK+ compared to controls and was associated with a two-fold increase in random cell migration of PID-transduced LSK+ cells in transwell migration assays. PID-expressing LSK+ cells also demonstrated abnormal lamellipodia associated with significant increases in both cell surface area and cell perimeter. Because cytoskeletal changes may be linked to alterations in cell growth, we next examined the effect of Pak inhibition on cell survival and proliferation. PID-expressing LSK+ cells had decreased proliferation (17.7% vs 36.8% of cells in S-phase, p 〈 0.05) and increased apoptosis (48.1% vs 16.7% AnnexinV+ cells, p 〈 0.05) when compared to controls, respectively. These phenotypic changes were associated with decreased pERK and pAKT in PID-expressing LSK+. To confirm the importance of Pak activation of these proteins in HSC/P, we performed experiments to rescue the observed engraftment defect by co-transducing PID or Mieg3 with a constitutively active-ERK (ca-MEK1) or ca-AKT. We found ca-MEK1, but not ca-AKT, was able to increase proliferation in vitro (% proliferating cells for PID + empty vector = 6.1% and PID+ ca-MEK1 = 9.5%; p 〈 0.05) and partially but only transiently rescue Pak-deficient HSC/P engraftment (% donor cells for LSK+ transduced with: PID + empty vector =1.5%, PID + ca-MEK1 =15.8%, and PID + ca-AKT =0.5% at 3-weeks post-BMT; p 〈 0.05 for empty vector vs ca-MEK1). Finally, to determine which PakA pathway is critical in HSC engraftment we studied Pak genetic knock-out cells. We found that Pak2Δ/Δ -but not Pak1−/− -cells resulted in a profound HSC/P engraftment defect (% Pak2Δ/Δ vs Pak2flox/flox and Pak1−/− vs Pak1wt/wt: 1.0% vs 26.5% and 35.8% vs 37.4%; p 〈 0.05 and p=ns, respectively at 3-weeks). Taken together, these data suggest that Pak A proteins regulate multiple HSC/P functions and link Rac GTPases to actin cytoskeletal rearrangements, directed cell migration, and proliferation/survival of HSC/P during engraftment.TABLE 1:Percentage of GFP+ cells in peripheral blood of recipient mice at indicated time points post BMT3-weeks6-weeks10-weeks14-weeks24-weeksWT-Mieg331.6% (±6.69)27.1% (±8.6)34.6% (±15.0)43.2% (±16.3)22.2% (±10.7)WT-PID0.22 (±0.19)0.07% (0.06)0%0%0%**Data represent mean ± s.d., n=10 recipients per group, p 〈 0.05 for all time points, two independent experiments. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 121, No. 13 ( 2013-03-28), p. 2474-2482
    Abstract: The Rac effector p21-activated kinase (Pak) regulates hematopoietic engraftment. Pak integrates cytoskeletal changes and proliferation pathways.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 125, No. 19 ( 2015-05-07), p. 2995-3005
    Abstract: Bone marrow-specific deletion of Pak2 is associated with macrothrombocytopenia and abnormal megakaryocyte morphology and function. Pak2 deletion is associated with defects in megakaryocyte endomitosis and the activation of Aurora-A and LIM kinase.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    H1 Connect ; 2010
    In:  F1000 Biology Reports Vol. 2 ( 2010-4-12)
    In: F1000 Biology Reports, H1 Connect, Vol. 2 ( 2010-4-12)
    Type of Medium: Online Resource
    ISSN: 1757-594X
    Language: Unknown
    Publisher: H1 Connect
    Publication Date: 2010
    detail.hit.zdb_id: 2486077-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...