Network


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

Hotspot


Dive into the research topics where Carolina L. Bigarella is active.

Publication


Featured researches published by Carolina L. Bigarella.


BMC Biology | 2010

Adherens junctions connect stress fibres between adjacent endothelial cells

Jaime Millán; Robert J. Cain; Natalia Reglero-Real; Carolina L. Bigarella; Beatriz Marcos-Ramiro; Laura Fernández-Martín; Isabel Correas; Anne J. Ridley

BackgroundEndothelial cell-cell junctions maintain endothelial integrity and regulate vascular morphogenesis and homeostasis. Cell-cell junctions are usually depicted with a linear morphology along the boundaries between adjacent cells and in contact with cortical F-actin. However, in the endothelium, cell-cell junctions are highly dynamic and morphologically heterogeneous.ResultsWe report that endothelial cell-cell junctions can attach to the ends of stress fibres instead of to cortical F-actin, forming structures that we name discontinuous adherens junctions (AJ). Discontinuous AJ are highly dynamic and are increased in response to tumour necrosis factor (TNF)-α, correlating with the appearance of stress fibres. We show that vascular endothelial (VE)-cadherin/β-catenin/α-catenin complexes in discontinuous AJ are linked to stress fibres. Moreover, discontinuous AJ connect stress fibres from adjacent cells independently of focal adhesions, of which there are very few in confluent endothelial cells, even in TNF-α-stimulated cells. RNAi-mediated knockdown of VE-cadherin, but not zonula occludens-1, reduces the linkage of stress fibres to cell-cell junctions, increases focal adhesions, and dramatically alters the distribution of these actin cables in confluent endothelial cells.ConclusionsOur results indicate that stress fibres from neighbouring cells are physically connected through discontinuous AJ, and that stress fibres can be stabilized by AJ-associated multi-protein complexes distinct from focal adhesions.


Development | 2014

Stem cells and the impact of ROS signaling.

Carolina L. Bigarella; Raymond Liang; Saghi Ghaffari

An appropriate balance between self-renewal and differentiation is crucial for stem cell function during both early development and tissue homeostasis throughout life. Recent evidence from both pluripotent embryonic and adult stem cell studies suggests that this balance is partly regulated by reactive oxygen species (ROS), which, in synchrony with metabolism, mediate the cellular redox state. In this Primer, we summarize what ROS are and how they are generated in the cell, as well as their downstream molecular targets. We then review recent findings that provide molecular insights into how ROS signaling can influence stem cell homeostasis and lineage commitment, and discuss the implications of this for reprogramming and stem cell ageing. We conclude that ROS signaling is an emerging key regulator of multiple stem cell populations.


Stem cell reports | 2014

Aging-like Phenotype and Defective Lineage Specification in SIRT1-Deleted Hematopoietic Stem and Progenitor Cells

Pauline Rimmele; Carolina L. Bigarella; Raymond Liang; Brigitte Izac; Rebeca Dieguez-Gonzalez; Gaetan Barbet; Michael J. Donovan; Carlo Brugnara; Julie Magarian Blander; David A. Sinclair; Saghi Ghaffari

Summary Aging hematopoietic stem cells (HSCs) exhibit defective lineage specification that is thought to be central to increased incidence of myeloid malignancies and compromised immune competence in the elderly. Mechanisms underlying these age-related defects remain largely unknown. We show that the deacetylase Sirtuin (SIRT)1 is required for homeostatic HSC maintenance. Differentiation of young SIRT1-deleted HSCs is skewed toward myeloid lineage associated with a significant decline in the lymphoid compartment, anemia, and altered expression of associated genes. Combined with HSC accumulation of damaged DNA and expression patterns of age-linked molecules, these have striking overlaps with aged HSCs. We further show that SIRT1 controls HSC homeostasis via the longevity transcription factor FOXO3. These findings suggest that SIRT1 is essential for HSC homeostasis and lineage specification. They also indicate that SIRT1 might contribute to delaying HSC aging.


Nature Immunology | 2015

CDKN1A regulates Langerhans cell survival and promotes Treg cell generation upon exposure to ionizing irradiation

Jeremy Price; Juliana Idoyaga; Hélène Salmon; Brandon Hogstad; Carolina L. Bigarella; Saghi Ghaffari; Marylene Leboeuf; Miriam Merad

Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a−/− LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a−/− LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.Treatment with ionizing irradiation (IR) may lead to accumulation of tumor-infiltrating T regulatory (Treg) cells and subsequent tumor resistance to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes, Langerhans cells (LCs), to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage post-IR. Particularly, we found that CDKN1A (cyclin-dependent kinase inhibitor 1A, also known as p21) was overexpressed in LCs, and that Cdkn1a−/− LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type, but not Cdkn1a−/−, LCs up-regulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and increased Treg cell numbers upon exposure to IR. These findings suggest a means for manipulating LC IR-resistance to increase cutaneous tumor response to radiotherapy.


EMBO Reports | 2015

Mitochondrial metabolism in hematopoietic stem cells requires functional FOXO3

Pauline Rimmele; Raymond Liang; Carolina L. Bigarella; Fatih Kocabas; Jingjing Xie; Madhavika N. Serasinghe; Jerry E. Chipuk; Hesham A. Sadek; Cheng Cheng Zhang; Saghi Ghaffari

Hematopoietic stem cells (HSC) are primarily dormant but have the potential to become highly active on demand to reconstitute blood. This requires a swift metabolic switch from glycolysis to mitochondrial oxidative phosphorylation. Maintenance of low levels of reactive oxygen species (ROS), a by‐product of mitochondrial metabolism, is also necessary for sustaining HSC dormancy. Little is known about mechanisms that integrate energy metabolism with hematopoietic stem cell homeostasis. Here, we identify the transcription factor FOXO3 as a new regulator of metabolic adaptation of HSC. ROS are elevated in Foxo3−/− HSC that are defective in their activity. We show that Foxo3−/− HSC are impaired in mitochondrial metabolism independent of ROS levels. These defects are associated with altered expression of mitochondrial/metabolic genes in Foxo3−/− hematopoietic stem and progenitor cells (HSPC). We further show that defects of Foxo3−/− HSC long‐term repopulation activity are independent of ROS or mTOR signaling. Our results point to FOXO3 as a potential node that couples mitochondrial metabolism with HSC homeostasis. These findings have critical implications for mechanisms that promote malignant transformation and aging of blood stem and progenitor cells.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Crosstalk Between Reticular Adherens Junctions and Platelet Endothelial Cell Adhesion Molecule-1 Regulates Endothelial Barrier Function

Laura Fernández-Martín; Beatriz Marcos-Ramiro; Carolina L. Bigarella; Mariona Graupera; Robert J. Cain; Natalia Reglero-Real; Anaïs Jiménez; Eva Cernuda-Morollón; Isabel Correas; Susan Cox; Anne J. Ridley; Jaime Millán

Objective—Endothelial cells provide a barrier between the blood and tissues, which is reduced during inflammation to allow selective passage of molecules and cells. Adherens junctions (AJ) play a central role in regulating this barrier. We aim to investigate the role of a distinctive 3-dimensional reticular network of AJ found in the endothelium. Methods and Results—In endothelial AJ, vascular endothelial-cadherin recruits the cytoplasmic proteins &bgr;-catenin and p120-catenin. &bgr;-catenin binds to &agr;-catenin, which links AJ to actin filaments. AJ are usually described as linear structures along the actin-rich intercellular contacts. Here, we show that these AJ components can also be organized in reticular domains that contain low levels of actin. Reticular AJ are localized in areas where neighboring cells overlap and encompass the cell adhesion receptor platelet endothelial cell adhesion molecule-1 (PECAM-1). Superresolution microscopy revealed that PECAM-1 forms discrete structures distinct from and distributed along AJ, within the voids of reticular domains. Inflammatory tumor necrosis factor-&agr; increases permeability by mechanisms that are independent of actomyosin-mediated tension and remain incompletely understood. Reticular AJ, but not actin-rich linear AJ, were disorganized by tumor necrosis factor-&agr;. This correlated with PECAM-1 dispersal from cell borders. PECAM-1 inhibition with blocking antibodies or small interfering RNA specifically disrupted reticular AJ, leaving linear AJ intact. This disruption recapitulated typical tumor necrosis factor-&agr;–induced alterations of barrier function, including increased &bgr;-catenin phosphorylation, without altering the actomyosin cytoskeleton. Conclusion—We propose that reticular AJ act coordinately with PECAM-1 to maintain endothelial barrier function in regions of low actomyosin-mediated tension. Selective disruption of reticular AJ contributes to permeability increase in response to tumor necrosis factor-&agr;.


PLOS Genetics | 2015

A Systems Approach Identifies Essential FOXO3 Functions at Key Steps of Terminal Erythropoiesis

Raymond Liang; Genís Campreciós; Yan Kou; Kathleen E. McGrath; Roberta B. Nowak; Seana C. Catherman; Carolina L. Bigarella; Pauline Rimmele; Xin Zhang; Merlin Nithya Gnanapragasam; James J. Bieker; Dmitri Papatsenko; Avi Ma’ayan; Emery H. Bresnick; Velia M. Fowler; James Palis; Saghi Ghaffari

Circulating red blood cells (RBCs) are essential for tissue oxygenation and homeostasis. Defective terminal erythropoiesis contributes to decreased generation of RBCs in many disorders. Specifically, ineffective nuclear expulsion (enucleation) during terminal maturation is an obstacle to therapeutic RBC production in vitro. To obtain mechanistic insights into terminal erythropoiesis we focused on FOXO3, a transcription factor implicated in erythroid disorders. Using an integrated computational and experimental systems biology approach, we show that FOXO3 is essential for the correct temporal gene expression during terminal erythropoiesis. We demonstrate that the FOXO3-dependent genetic network has critical physiological functions at key steps of terminal erythropoiesis including enucleation and mitochondrial clearance processes. FOXO3 loss deregulated transcription of genes implicated in cell polarity, nucleosome assembly and DNA packaging-related processes and compromised erythroid enucleation. Using high-resolution confocal microscopy and imaging flow cytometry we show that cell polarization is impaired leading to multilobulated Foxo3 -/- erythroblasts defective in nuclear expulsion. Ectopic FOXO3 expression rescued Foxo3 -/- erythroblast enucleation-related gene transcription, enucleation defects and terminal maturation. Remarkably, FOXO3 ectopic expression increased wild type erythroblast maturation and enucleation suggesting that enhancing FOXO3 activity may improve RBCs production. Altogether these studies uncover FOXO3 as a novel regulator of erythroblast enucleation and terminal maturation suggesting FOXO3 modulation might be therapeutic in disorders with defective erythroid maturation.


Journal of Biological Chemistry | 2013

ARHGAP21 Protein, a New Partner of α-Tubulin Involved in Cell-Cell Adhesion Formation and Essential for Epithelial-Mesenchymal Transition

Karin Spat Albino Barcellos; Carolina L. Bigarella; Mark V. Wagner; Karla Priscila Vieira; Mariana Lazarini; Peter R. Langford; João Agostinho Machado-Neto; Steven G. Call; Davis M. Staley; Jarom Y. Chung; Marc D.H. Hansen; Sara Teresinha Olalla Saad

Background: ARHGAP21 is an important Rho-GAP for Cdc42 involved in vesicle trafficking and focal adhesion kinase activity. Results: ARHGAP21 participates in cell-cell adhesion formation and cellular migration, interacts and modulates α-tubulin acetylation, and is essential for epithelial-mesenchymal transition. Conclusion: ARHGAP21 is a novel α-tubulin partner coordinating cell-cell adhesion, migration, and epithelial-mesenchymal transition. Significance: ARHGAP21 might be involved in cancer metastasis. Cell-cell adhesions and the cytoskeletons play important and coordinated roles in cell biology, including cell differentiation, development, and migration. Adhesion and cytoskeletal dynamics are regulated by Rho-GTPases. ARHGAP21 is a negative regulator of Rho-GTPases, particularly Cdc42. Here we assess the function of ARHGAP21 in cell-cell adhesion, cell migration, and scattering. We find that ARHGAP21 is localized in the nucleus, cytoplasm, or perinuclear region but is transiently redistributed to cell-cell junctions 4 h after initiation of cell-cell adhesion. ARHGAP21 interacts with Cdc42, and decreased Cdc42 activity coincides with the appearance of ARHGAP21 at the cell-cell junctions. Cells lacking ARHGAP21 expression show weaker cell-cell adhesions, increased cell migration, and a diminished ability to undergo hepatocyte growth factor-induced epithelial-mesenchymal transition (EMT). In addition, ARHGAP21 interacts with α-tubulin, and it is essential for α-tubulin acetylation in EMT. Our findings indicate that ARHGAP21 is a Rho-GAP involved in cell-cell junction remodeling and that ARHGAP21 affects migration and EMT through α-tubulin interaction and acetylation.


Biochimica et Biophysica Acta | 2009

ARHGAP21 modulates FAK activity and impairs glioblastoma cell migration.

Carolina L. Bigarella; Luciene Rodrigues de Oliveira Borges; Fernando Ferreira Costa; Sara Terezinha Olalla Saad

Glioblastoma multiforme is highly aggressive and is the most common glial tumor type. Although there have been advances in treatment, the average survival expectancy is 12-15 months. Several genes have been shown to influence glioblastoma progression. In the present work, we demonstrate that the RhoGTPase Activating Protein 21 (ARHGAP21) is expressed in the nuclear and perinuclear regions of several cell lines. In T98G and U138MG, glioblastoma derived cell lines, ARHGAP21 interacts with the C-terminal region of Focal Adhesion Kinase (FAK). ARHGAP21 depletion by shRNAi in T98G cells alters cellular morphology and increases: FAK phosphorylation states and activation of downstream signaling; the activity state of Cdc42; the production of metalloproteinase 2 (MMP-2) and cell migration rates. These modifications were found to be mainly due to the loss of ARHGAP21 action on FAK and, consequently, the activation of downstream effectors. These results suggest not only that ARHGAP21 might act as a tumor suppressor gene, but also indicate that ARHGAP21 might be a master regulator of migration having a crucial role in controlling the progression of different tumor types.


PLOS ONE | 2014

CXCR7 is highly expressed in acute lymphoblastic leukemia and potentiates CXCR4 response to CXCL12.

Rita de Cássia Carvalho Melo; Ana Leda Longhini; Carolina L. Bigarella; Mariana Ozello Baratti; Fabiola Traina; Patricia Favaro; Paula de Melo Campos; Sara Teresinha Olalla Saad

Recently, a novel CXCL12-binding receptor, has been identified. This CXCL12-binding receptor commonly known as CXCR7 (CXC chemokine receptor 7), has lately, based on a novel nomenclature, has received the name ACKR3 (atypical chemokine receptor 3). In this study, we aimed to investigate the expression of CXCR7 in leukemic cells, as well as its participation in CXCL12 response. Interesting, we clearly demonstrated that CXCR7 is highly expressed in acute lymphoid leukemic cells compared with myeloid or normal hematopoietic cells and that CXCR7 contributed to T-acute lymphoid leukemic cell migration induced by CXCL12. Moreover, we showed that the cellular location of CXCR7 varied among T-lymphoid cells and this finding may be related to their migration capacity. Finally, we hypothesized that CXCR7 potentiates CXCR4 response and may contribute to the maintenance of leukemia by initiating cell recruitment to bone marrow niches that were once occupied by normal hematopoietic stem cells.

Collaboration


Dive into the Carolina L. Bigarella's collaboration.

Top Co-Authors

Avatar

Saghi Ghaffari

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pauline Rimmele

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Raymond Liang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Fabiola Traina

University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Brigitte Izac

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Valentina d'Escamard

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Mariana Lazarini

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge