Network


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

Hotspot


Dive into the research topics where Tina M. Leisner is active.

Publication


Featured researches published by Tina M. Leisner.


Journal of Cell Biology | 2005

Essential role of CIB1 in regulating PAK1 activation and cell migration

Tina M. Leisner; Mingjuan Liu; Zahara M. Jaffer; Jonathan Chernoff; Leslie V. Parise

p21-activated kinases (PAKs) regulate many cellular processes, including cytoskeletal rearrangement and cell migration. In this study, we report a direct and specific interaction of PAK1 with a 22-kD Ca2+-binding protein, CIB1, which results in PAK1 activation both in vitro and in vivo. CIB1 binds to PAK1 within discrete regions surrounding the inhibitory switch domain in a calcium-dependent manner, providing a potential mechanism of CIB1-induced PAK1 activation. CIB1 overexpression significantly decreases cell migration on fibronectin as a result of a PAK1-and LIM kinase–dependent increase in cofilin phosphorylation. Conversely, the RNA interference–mediated depletion of CIB1 increases cell migration and reduces normal adhesion-induced PAK1 activation and cofilin phosphorylation. Together, these results demonstrate that endogenous CIB1 is required for regulated adhesion-induced PAK1 activation and preferentially induces a PAK1-dependent pathway that can negatively regulate cell migration. These results point to CIB1 as a key regulator of PAK1 activation and signaling.


Journal of Cell Biology | 2006

CIB1 is an endogenous inhibitor of agonist-induced integrin αIIbβ3 activation

Weiping Yuan; Tina M. Leisner; Andrew W. McFadden; Zhengyan Wang; Mark K. Larson; Shantres Clark; Christel Boudignon-Proudhon; Stephen C.-T. Lam; Leslie V. Parise

In response to agonist stimulation, the αIIbβ3 integrin on platelets is converted to an active conformation that binds fibrinogen and mediates platelet aggregation. This process contributes to both normal hemostasis and thrombosis. Activation of αIIbβ3 is believed to occur in part via engagement of the β3 cytoplasmic tail with talin; however, the role of the αIIb tail and its potential binding partners in regulating αIIbβ3 activation is less clear. We report that calcium and integrin binding protein 1 (CIB1), which interacts directly with the αIIb tail, is an endogenous inhibitor of αIIbβ3 activation; overexpression of CIB1 in megakaryocytes blocks agonist-induced αIIbβ3 activation, whereas reduction of endogenous CIB1 via RNA interference enhances activation. CIB1 appears to inhibit integrin activation by competing with talin for binding to αIIbβ3, thus providing a model for tightly controlled regulation of αIIbβ3 activation.


Molecular and Cellular Biology | 2006

CIB1 is essential for mouse spermatogenesis.

Weiping Yuan; Tina M. Leisner; Andrew W. McFadden; Shantres Clark; Sylvia Hiller; Nobuyo Maeda; Deborah A. O'Brien; Leslie V. Parise

ABSTRACT CIB1 is a 22-kDa calcium binding, regulatory protein with ∼50% homology to calmodulin and calcineurin B. CIB1 is widely expressed and binds to a number of effectors, such as integrin αIIb, PAK1, and polo-like kinases, in different tissues. However, the in vivo functions of CIB1 are not well understood. To elucidate the function of CIB1 in whole animals, we used homologous recombination in embryonic stem cells to generate Cib1−/− mice. Although Cib1−/− mice grow normally, the males are sterile due to disruption of the haploid phase of spermatogenesis. This is associated with reduced testis size and numbers of germ cells in seminiferous tubules, increased germ cell apoptosis, and the loss of elongated spermatids and sperm. Cib1−/− testes also show increased mRNA and protein expression of the cell cycle regulator Cdc2/Cdk1. In addition, mouse embryonic fibroblasts (MEFs) derived from Cib1−/− mice exhibit a much slower growth rate compared to Cib1+/+ MEFs, suggesting that CIB1 regulates the cell cycle, differentiation of spermatogenic germ cells, and/or differentiation of supporting Sertoli cells.


Oncogene | 2013

CIB1 prevents nuclear GAPDH accumulation and non-apoptotic tumor cell death via AKT and ERK signaling

Tina M. Leisner; Cassandra Moran; Stephen P. Holly; Leslie V. Parise

CIB1 is a 22-kDa regulatory protein previously implicated in cell survival and proliferation. However, the mechanism by which CIB1 regulates these processes is poorly defined. Here, we report that CIB1 depletion in SK-N-SH neuroblastoma and MDA-MB-468 breast cancer cells promotes non-apoptotic, caspase-independent cell death that is not initiated by increased outer mitochondrial membrane permeability or translocation of apoptosis-inducing factor to the nucleus. Instead, cell death requires nuclear GAPDH accumulation. Furthermore, CIB1 depletion disrupts two commonly dysregulated, oncogenic pathways—PI3K/AKT and Ras/MEK/ERK, resulting in a synergistic mechanism of cell death, which was mimicked by simultaneous pharmacological inhibition of both pathways, but not either pathway alone. In defining each pathway’s contributions, we found that AKT inhibition alone maximally induced GAPDH nuclear accumulation, whereas MEK/ERK inhibition alone had no effect on GAPDH localization. Concurrent GAPDH nuclear accumulation and ERK inhibition were required, however, to induce a significant DNA damage response, which was critical to subsequent cell death. Collectively, our results indicate that CIB1 is uniquely positioned to regulate PI3K/AKT and MEK/ERK signaling and that simultaneous disruption of these pathways synergistically induces a nuclear GAPDH-dependent cell death. The mechanistic insights into cell death induced by CIB1 interference suggest novel molecular targets for cancer therapy.


Circulation Research | 2007

CIB1 Regulates Endothelial Cells and Ischemia-Induced Pathological and Adaptive Angiogenesis

Mohamed A. Zayed; Weiping Yuan; Tina M. Leisner; Dan Chalothorn; Andrew W. McFadden; Michael D. Schaller; M. Elizabeth Hartnett; James E. Faber; Leslie V. Parise

Pathological angiogenesis contributes to various ocular, malignant, and inflammatory disorders, emphasizing the need to understand this process on a molecular level. CIB1 (calcium- and integrin-binding protein), a 22-kDa EF-hand–containing protein, modulates the activity of p21-activated kinase 1 in fibroblasts. Because p21-activated kinase 1 also contributes to endothelial cell function, we hypothesized that CIB1 may have a role in angiogenesis. We found that endothelial cells depleted of CIB1 by either short hairpin RNA or homologous recombination have reduced migration, proliferation, and tubule formation. Moreover, loss of CIB1 in these cells decreases p21-activated kinase 1 activation, downstream extracellular signal-regulated kinase 1/2 activation, and matrix metalloproteinase 2 expression, all of which are known to contribute to angiogenesis. Consistent with these findings, tissues derived from CIB1-deficient (CIB1−/−) mice have reduced growth factor–induced microvessel sprouting in ex vivo organ cultures and in vivo Matrigel plugs. Furthermore, in response to ischemia, CIB1−/− mice demonstrate decreased pathological retinal and adaptive hindlimb angiogenesis. Ischemic CIB1−/− hindlimbs also demonstrate increased tissue damage and significantly reduced p21-activated kinase 1 activation. These data therefore reveal a critical role for CIB1 in ischemia-induced pathological and adaptive angiogenesis.


Current Opinion in Hematology | 2007

Tickling the tails: Cytoplasmic domain proteins that regulate integrin αIIbβ3 activation

Tina M. Leisner; Weiping Yuan; Jan C. DeNofrio; Jun Liu; Leslie V. Parise

Purpose of reviewIntegrin αIIbβ3 activation is essential for platelet aggregation and related hemostatic events. In recent years, intense effort has been put forward to understand the molecular mechanisms regulating platelet integrin αIIbβ3 activation. Here we review the current models of αIIbβ3 activation and highlight the potential regulatory roles of proteins that interact directly with the αIIbβ3 cytoplasmic domains, with emphasis on the αIIb cytoplasmic domain binding protein, CIB1. Recent findingsMutational and crystallographic studies reveal the importance of integrin transmembrane and cytoplasmic domains in propagating bidirectional signaling events. Proteins that interact directly with the integrin cytoplasmic domains may play important roles in mediating these signaling events. Of particular interest is the interaction between CIB1 and the αIIb tail which may function to negatively regulate αIIbβ3 activation. In addition, a number of CIB1 interacting proteins have been identified, including p21-activated kinase and serum-inducible kinase, which may act in concert with CIB1 to regulate platelet function. SummaryUnderstanding the molecular mechanisms underlying integrin activation will be important in developing novel therapies to regulate platelet function in cardiovascular disease. Discussion of recent developments in elucidating the mechanism of integrin activation, with particular focus on the platelet integrin αIIbβ3, is provided in this review.


The FASEB Journal | 2016

CIB1: a small protein with big ambitions.

Tina M. Leisner; Thomas C. Freeman; Justin L. Black; Leslie V. Parise

Calcium‐ and integrin‐binding protein 1 (CIB1) is a small, ubiquitously expressed protein that was first identified as an intracellular binding partner of a platelet‐specific α‐integrin cytoplasmic tail. Although early studies revealed a role for CIB1 in regulating platelet integrin activity, recent studies have indicated a more diverse role for CIB1 in many different cell types and processes, including calcium signaling, migration, adhesion, proliferation, and survival. Increasing evidence also points to a novel role for CIB1 in cancer and cardiovascular disease. In addition, an array of CIB1 binding partners has been identified that provide important insight into how CIB1 may regulate these processes. Some of these binding partners include the serine/threonine kinases, p21‐activated kinase 1 (PAK1), apoptosis signal‐regulating kinase 1 (ASK1), and polo‐like kinase 3 (PLK3). Structural and mutational studies indicate that CIB1 binds most or all of its partners via a well‐defined hydrophobic cleft. Although CIB1 itself lacks known enzymatic activity, it supports the PI3K/AKT and MEK/ERK oncogenic signaling pathways, in part, by directly modulating enzymes in these pathways. In this review, we discuss our current understanding of CIB1 and key questions regarding structure and function and how this seemingly diminutive protein impacts important signaling pathways and cellular processes in human health and disease.—Leisner, T. M., Freeman, T. C., Black, J. L., Parise, L. V. CIB1: a small protein with big ambitions. FASEB J. 30, 2640‐2650 (2016). www.fasebj.org


Biochemistry | 2013

Identification of Novel Integrin Binding Partners for Calcium and Integrin Binding Protein 1 (CIB1): Structural and Thermodynamic Basis of CIB1 Promiscuity

Thomas C. Freeman; Justin L. Black; Holly G. Bray; Onur Dagliyan; Yi I. Wu; Ashutosh Tripathy; Nikolay V. Dokholyan; Tina M. Leisner; Leslie V. Parise

The short cytoplasmic tails of the α- and β-chains of integrin adhesion receptors regulate integrin activation and cell signaling. Significantly less is known about proteins that bind to α-integrin cytoplasmic tails (CTs) as opposed to β-CTs to regulate integrins. Calcium and integrin binding protein 1 (CIB1) was previously identified as an αIIb binding partner that inhibits agonist-induced activation of the platelet-specific integrin, αIIbβ3. A sequence alignment of all α-integrin CTs revealed that key residues in the CIB1 binding site of αIIb are well-conserved, and was used to delineate a consensus binding site (I/L-x-x-x-L/M-W/Y-K-x-G-F-F). Because the CIB1 binding site of αIIb is conserved in all α-integrins and CIB1 expression is ubiquitous, we asked if CIB1 could interact with other α-integrin CTs. We predicted that multiple α-integrin CTs were capable of binding to the same hydrophobic binding pocket on CIB1 with docking models generated by all-atom replica exchange discrete molecular dynamics. After demonstrating novel in vivo interactions between CIB1 and other whole integrin complexes with co-immunoprecipitations, we validated the modeled predictions with solid-phase competitive binding assays, which showed that other α-integrin CTs compete with the αIIb CT for binding to CIB1 in vitro. Isothermal titration calorimetry measurements indicated that this binding is driven by hydrophobic interactions and depends on residues in the CIB1 consensus binding site. These new mechanistic details of CIB1-integrin binding imply that CIB1 could bind to all integrin complexes and act as a broad regulator of integrin function.


Breast Cancer Research and Treatment | 2015

CIB1 depletion impairs cell survival and tumor growth in triple-negative breast cancer.

Justin L. Black; J. Chuck Harrell; Tina M. Leisner; Melissa J. Fellmeth; Samuel D. George; Dominik Reinhold; Nicole M. Baker; Corbin D. Jones; Channing J. Der; Charles M. Perou; Leslie V. Parise

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with generally poor prognosis and no available targeted therapies, highlighting a critical unmet need to identify and characterize novel therapeutic targets. We previously demonstrated that CIB1 is necessary for cancer cell survival and proliferation via regulation of two oncogenic signaling pathways, RAF–MEK–ERK and PI3K–AKT. Because these pathways are often upregulated in TNBC, we hypothesized that CIB1 may play a broader role in TNBC cell survival and tumor growth. Methods utilized include inducible RNAi depletion of CIB1 in vitro and in vivo, immunoblotting, clonogenic assay, flow cytometry, RNA-sequencing, bioinformatics analysis, and Kaplan–Meier survival analysis. CIB1 depletion resulted in significant cell death in 8 of 11 TNBC cell lines tested. Analysis of components related to PI3K–AKT and RAF–MEK–ERK signaling revealed that elevated AKT activation status and low PTEN expression were key predictors of sensitivity to CIB1 depletion. Furthermore, CIB1 knockdown caused dramatic shrinkage of MDA-MB-468 xenograft tumors in vivo. RNA sequence analysis also showed that CIB1 depletion in TNBC cells activates gene programs associated with decreased proliferation and increased cell death. CIB1 expression levels per se did not predict TNBC susceptibility to CIB1 depletion, and CIB1 mRNA expression levels did not associate with TNBC patient survival. Our data are consistent with the emerging concept of non-oncogene addiction, where a large subset of TNBCs depend on CIB1 for cell survival and tumor growth, independent of CIB1 expression levels. Our data establish CIB1 as a novel therapeutic target for TNBC.


Blood | 2011

Talin's second act-ivation: retraction

Tina M. Leisner; Leslie V. Parise

In this issue of Blood, Haling and colleagues demonstrate that in addition to talin-dependent integrin activation, talin is required for platelet fibrin clot retraction by physically linking integrins to the actin cytoskeleton.

Collaboration


Dive into the Tina M. Leisner's collaboration.

Top Co-Authors

Avatar

Leslie V. Parise

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Weiping Yuan

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Andrew W. McFadden

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Justin L. Black

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Shantres Clark

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Zhengyan Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ashutosh Tripathy

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Christel Boudignon-Proudhon

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Holly G. Bray

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Nikolay V. Dokholyan

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge