Network


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

Hotspot


Dive into the research topics where Carla Motta is active.

Publication


Featured researches published by Carla Motta.


Journal of Lipid Research | 2007

Endothelial cell-pericyte cocultures induce PLA2 protein expression through activation of PKCα and the MAPK/ERK cascade

Carmelina Daniela Anfuso; Gabriella Lupo; Loriana Romeo; Giovanni Giurdanella; Carla Motta; Alessia Pascale; Cataldo Tirolo; Bianca Marchetti; Mario Alberghina

Little is known about the regulatory mechanisms of endothelial cell (EC) proliferation by retinal pericytes and vice versa. In a model of coculture with bovine retinal pericytes lasting for 24 h, rat brain ECs showed an increase in arachidonic acid (AA) release, whereas Western blot and RT-PCR analyses revealed that ECs activated the protein expression of cytosolic phospholipase A2 (cPLA2) and its phosphorylated form and calcium-independent intracellular phospholipase A2 (iPLA2). No activation of the same enzymes was seen in companion pericytes. In ECs, the protein level of phosphorylated extracellular signal-regulated kinase (ERK) 1/2 was also enhanced significantly, a finding not observed in cocultured pericytes. The expression of protein kinase C-α (PKCα) and its phosphorylated form was also enhanced in ECs. Wortmannin, LY294002, and PD98059, used as inhibitors of upstream kinases (the PI3-kinase/Akt/PDK1 or MEK-1 pathway) in cultures, markedly attenuated AA release and the expression of phosphorylated forms of endothelial cPLA2, PKCα, and ERK1/2. By confocal microscopy, activation of PKCα in perinuclear regions of ECs grown in coculture as well as strong activation of cPLA2 in ECs taken from a model of mixed culture were clearly observed. However, no increased expression of both enzymes was found in cocultured pericytes. Our findings indicate that a sequential activation of PKCα contributes to endothelial ERK1/2 and cPLA2 phosphorylation induced by either soluble factors or direct cell-to-cell contact, and that the PKCα-cPLA2 pathway appears to play a key role in the early phase of EC-pericyte interactions regulating blood retina or blood-brain barrier maturation.


Biochemical Pharmacology | 2013

Role of phospholipases A2 in diabetic retinopathy: In vitro and in vivo studies

Gabriella Lupo; Carla Motta; Giovanni Giurdanella; Carmelina Daniela Anfuso; Mario Alberghina; Filippo Drago; Salvatore Salomone; Claudio Bucolo

Diabetic retinopathy is one of the leading causes of blindness and the most common complication of diabetes with no cure available. We investigated the role of phospholipases A2 (PLA2) in diabetic retinopathy using an in vitro blood-retinal barrier model (BRB) and an in vivo streptozotocin (STZ)-induced diabetic model. Mono- and co-cultures of endothelial cells (EC) and pericytes (PC), treated with high or fluctuating concentrations of glucose, to mimic the diabetic condition, were used. PLA2 activity, VEGF and PGE2 levels and cell proliferation were measured, with or without PLA2 inhibition. Diabetes was induced in rats by STZ injection and PLA2 activity along with VEGF, TNFα and ICAM-1 levels were measured in retina. High or fluctuating glucose induced BRB breakdown, and increased PLA2 activity, PGE2 and VEGF in EC/PC co-cultures; inhibition of PLA2 in mono- or co-cultures treated with high or fluctuating glucose dampened PGE2 and VEGF production down to the levels of controls. High or fluctuating glucose increased EC number and reduced PC number in co-cultures; these effects were reversed after transfecting EC with small interfering RNA targeted to PLA2. PLA2 and COX-2 protein expressions were significantly increased in microvessels from retina of diabetic rats. Diabetic rats had also high retinal levels of VEGF, ICAM-1 and TNFα that were reduced by treatment with a cPLA2 inhibitor. In conclusion, the present findings indicate that PLA2 upregulation represents an early step in glucose-induced alteration of BRB, possibly upstream of VEGF; thus, PLA2 may be an interesting target in managing diabetic retinopathy.


Biochimica et Biophysica Acta | 2008

Expression of Ca2+-independent and Ca2+-dependent phospholipases A2 and cyclooxygenases in human melanocytes and malignant melanoma cell lines

Mariagrazia Rita Scuderi; Carmelina Daniela Anfuso; Gabriella Lupo; Carla Motta; Loriana Romeo; Liliana Guerra; Alessandro Cappellani; Nicola Ragusa; Giuseppina Cantarella; Mario Alberghina

We provide novel evidence that human melanoma cell lines (M10, M14, SK-MEL28, SK-MEL93, 243MEL, 1074MEL, OCM-1, and COLO38) expressed, at mRNA and protein levels, either Ca(2+)-independent phospholipase A(2) (iPLA(2)) or cytosolic phospholipase A(2) (cPLA(2)) and its phosphorylated form. Normal human melanocytes contained the lowest levels of both PLA(2)s. Cyclooxygenase-1 and -2 (COX-1 and COX-2) were also expressed in cultured tumor cells as measured by Western blots. The most pronounced overexpression of iPLA(2) and COX-1 was found in two melanoma-derived cells, M14 and COLO38. Normal human melanocytes and the M10 melanoma cell line displayed no COX-2 expression. Using subcellular fractionation, Western blot and confocal microcopy analyses, in paradigmatic SK-MEL28 and SK-MEL93 cells we showed that iPLA(2), COX-1 and even cPLA(2) were equally located in the cytosol, membrane structures and perinuclear region while COX-2 was preferentially associated with the cytosol. Specific inhibitors of these three enzymes significantly reduced the basal proliferation rate either in melanocytes or in melanoma cell lines. These results, coupled with the inhibition of the cell proliferation by electroporation of melanoma cells with cPLA(2) or COX-2 antibodies, demonstrate that a possible correlation between PLA(2)-COX expression and tumor cell proliferation in the melanocytic system does exist. In addition, the high expression level of both PLA(2)s and COXs suggests that eicosanoids modulate cell proliferation and tumor invasiveness.


Microvascular Research | 2011

Cytosolic and calcium-independent phospholipase A2 mediate glioma-enhanced proangiogenic activity of brain endothelial cells

Giovanni Giurdanella; Carla Motta; Stefano Muriana; Valeria Arena; Carmelina Daniela Anfuso; Gabriella Lupo; Mario Alberghina

Glioma is characterized by an active production of proangiogenic molecules. We observed that conditioned medium (CM) from C6 glioma significantly enhanced proliferation and migration of immortalized rat brain GP8.3 endothelial cells (ECs) and primary bovine brain microvascular ECs. The glioma CM effect was significantly reduced by cytosolic (cPLA(2)) and Ca(++)-independent (iPLA(2)) phospholipase A(2), cyclooxygenase-2, and protein kinase inhibitors. In GP8.3 ECs, cPLA(2) and iPLA(2) enzyme activities and phosphorylation of cPLA(2), significantly stimulated after 24h CM co-incubation, were attenuated by PLA(2), PI3-K, MEK-1, and ERK1/2 inhibitors. By confocal microscopy, in glioma CM-stimulated ECs, enhancement of fluorescence signals for phospho-cPLA(2), phospho-ERK1/2, phospho-PKCα, COX-2, and iPLA(2) was in parallel observed. Electroporation of anti-iPLA(2) and cPLA(2) antibodies and siRNAs directed against iPLA(2) and cPLA(2) significantly inhibited cell proliferation and migration. Incubation of CM- or VEGF peptide-stimulated ECs with antibodies against VEGF or VEGFR-1/-2 receptors strongly reduced mitotic rate, cell migration, and phospho-cPLA(2) and iPLA(2) protein levels. The findings suggest that PLA(2) activities are involved in stimulating EC migration and proliferation in the presence of glioma CM and that cPLA(2) is positively regulated upstream by PI3-K, PKCα, and ERK1/2 signal cascades. Our work provides new insights in understanding EC metabolism and signaling during tumor angiogenesis.


Cancer Letters | 2014

An in vitro retinoblastoma human triple culture model of angiogenesis: a modulatory effect of TGF-β.

Gabriella Lupo; Carla Motta; Mario Salmeri; Vittoria Spina-Purrello; Mario Alberghina; Carmelina Daniela Anfuso

Retinoblastoma is the most common intraocular tumour in children. In view of understanding the molecular mechanisms through which angiogenic switch on happens in the early phases of reciprocal interaction between tumour and cells constituting retinal microvessel, Transwell co-cultures constituted by human retinal endothelial cells (HREC), pericytes (HRPC), and human retinoblastoma cell line Y-79 were performed. Y-79 enhanced HREC proliferation, reduced by the introduction of HRPC in triple culture. In HREC/HRPC cultures, TGF-β in media increased, decreasing in triple cultures. High VEGF levels in triple cultures witnessed the establishment of a strongly in vitro angiogenic environment. Y-79 induced in HREC an increase in c- and iPLA2, phospho-cPLA2, inducible COX-2 protein expressions, PLA2 activities and prostaglandin E2 (PGE2) release. These effects were attenuated when HRPC were introduced in triple culture. Moreover, antibody silencing of TGF-β demonstrated a strong correlation between the signalling pathway triggered by TGF-β of pericytal origin and the phospholipase activation and the modulation of PGE2 release. Inhibiting VEGFA effect, the HRPC loss in triple culture decreased, showing its modulatory effect on their survival. Relying on the data here presented, sustaining the pericytal survival in a tumour retinal environment could ensure the integrity of microvessels and the TGF-β supply, essential for controlling aberrant endothelial pruning and angiogenesis.


Cellular Microbiology | 2013

VEGF receptor-1 involvement in pericyte loss induced by Escherichia coli in an in vitro model of blood brain barrier

Mario Salmeri; Carla Motta; Carmelina Daniela Anfuso; Andrea Amodeo; Marina Scalia; Maria Antonietta Toscano; Mario Alberghina; Gabriella Lupo

The key aspect of neonatal meningitis is related to the ability of pathogens to invade the blood–brain barrier (BBB) and to penetrate the central nervous system. In the present study we show that, in an in vitro model of BBB, on the basis of co‐culturing primary bovine brain endothelial cells (BBEC) and primary bovine retinal pericytes (BRPC), Escherichia coli infection determines changes of transendothelial electrical resistance (TEER) and permeability (Pe) to sodium fluorescein. In the co‐culture model, within BBEC, bacteria are able to stimulate cytosolic and Ca2+‐independent phospholipase A2 (cPLA2 and iPLA2) enzyme activities. In supernatants of E. coli‐stimulated co‐cultures, an increase in prostaglandins (PGE2) and VEGF production in comparison with untreated co‐cultures were found. Incubation with E. coli in presence of AACOCF3 or BEL caused a decrease of PGE2 and VEGF release. SEM and TEM images of BBEC and BRPC showed E. coli adhesion to BBEC and BRPC but only in BBEC the invasion occurs. VEGFR‐1 but not VEGFR‐2 blockade by the specific antibody reduced E. coli invasion in BBEC. In our model of BBB infection, a significant loss of BRPC was observed. Following VEGFR‐1, but not VEGFR‐2 blockade, or in presence of AACOCF3 or BEL, elevated TEER values, reducedpermeability and BRPC loss were found. These data suggest that VEGFR‐1 negatively regulates BRPC survival and its blockade protects the barrier integrity. PGs and VEGF could exert a biological effect on BBB, probably by BRPC coverage ablation, thus increasing BBB permeability. Our results show the role played by the BBEC as well as BRPC during a bacterial attack on BBB. A better understanding of the mechanisms by which E. coli enter the nervous system and how bacteria alter the communication between endothelial cells and pericytes may provide exciting new insight for clinical intervention.


Microvascular Research | 2009

PKCα-MAPK/ERK-phospholipase A2 signaling is required for human melanoma-enhanced brain endothelial cell proliferation and motility

Carmelina Daniela Anfuso; Giovanni Giurdanella; Carla Motta; Stefano Muriana; Gabriella Lupo; Nicola Ragusa; Mario Alberghina

The largely undefined signal transduction mechanisms and cross-talk between human melanoma cell (HMC) lines and brain endothelial cells (ECs) involved in tumor cell interaction and adhesion were investigated. In immortalized rat brain GP8.3 EC cultures, conditioned media (CM) prepared from SK-MEL28 and OCM-1 melanoma cells significantly enhanced arachidonic acid release, cytosolic phospholipase A(2) (cPLA(2)) and Ca(+)-independent phospholipase A(2) (iPLA(2)) specific activities, and cell growth by 24 h. Inhibitors such as wortmannin and LY294002 (vs. PI3 kinase activity), AACOCF(3), (vs. cPLA(2) and iPLA(2)), PD98059 (vs. ERK1/2 activity) and NS-398 (vs. cyclooxygenase-2 activity, COX-2) were all able to block cell proliferation and motility determined using a scratch wound healing assay in melanoma CMs-stimulated EC monolayers. These media also support the enhanced cell proliferation of primary ECs derived from rat brain (BBEC). Electroporation of anti-cPLA(2) antibody into ECs markedly inhibited the EC proliferation in response to CMs. With both CMs, phosphorylation of cPLA(2), PKCalpha, ERK1/2, protein and mRNA expression of cPLA(2) and iPLA(2), and COX-2 protein expression were significantly stimulated after 24 h coincubation, and attenuated by specific inhibitors. By confocal microscopy, activation of cPLA(2), ERK1/2, PKCalpha and COX-2 in perinuclear and membrane regions of ECs grown in CM-stimulated cultures were clearly observed. Thus MEK-PKCalpha-ERK1/2 and PI3-K/Akt survival pathways are activated in EC cultures during the interaction with CM from both melanoma cell lines, providing new insight in understanding EC metabolism and signaling. These pathways represent potential therapeutic targets to inhibit or enhance tumor angiogenesis.


Frontiers in Pharmacology | 2017

Anti-angiogenic Therapy in Cancer: Downsides and New Pivots for Precision Medicine

Gabriella Lupo; Nunzia Caporarello; Melania Olivieri; Martina Cristaldi; Carla Motta; Vincenzo Bramanti; Roberto Avola; Mario Salmeri; Ferdinando Nicoletti; Carmelina Daniela Anfuso

Primary solid tumors originate close to pre-existing tissue vasculature, initially growing along such tissue blood vessels, and this phenomenon is important for the metastatic potential which frequently occurs in highly vascularized tissues. Unfortunately, preclinic and clinic anti-angiogenic approaches have not been very successful, and multiple factors have been found to contribute to toxicity and tumor resistance. Moreover, tumors can highlight intrinsic or acquired resistances, or show adaptation to the VEGF-targeted therapies. Furthermore, different mechanisms of vascularization, activation of alternative signaling pathways, and increased tumor aggressiveness make this context even more complex. On the other hand, it has to be considered that the transitional restoration of normal, not fenestrated, microvessels allows the drug to reach the tumor and act with the maximum efficiency. However, these effects are time-limited and different, depending on the various types of cancer, and clearly define a specific “normalization window.” So, new horizons in the therapeutic approaches consist on the treatment of the tumor with pro- (instead of anti-) angiogenic therapies, which could strengthen a network of well-structured blood vessels that facilitate the transport of the drug.


Neuroscience Letters | 2012

Involvement of PKCα–MAPK/ERK-phospholipase A2 pathway in the Escherichia coli invasion of brain microvascular endothelial cells

Mario Salmeri; Carla Motta; Silvana Mastrojeni; Andrea Amodeo; Carmelina Daniela Anfuso; Giovanni Giurdanella; Angela Morello; Mario Alberghina; Maria Antonietta Toscano; Gabriella Lupo

Escherichia coli K1 is the most common Gram-negative organism that causes neonatal meningitis following penetration of the blood-brain barrier. In the present study we demonstrated the involvement of cytosolic (cPLA(2)) and calcium-independent phospholipase A(2) (iPLA(2)) and the contribution of cyclooxygenase-2 products in E. coli invasion of microvascular endothelial cells. The traversal of bacteria did not determine trans-endothelial electrical resistance (TEER) and ZO-1 expression changes and was reduced by PLA(2)s siRNA. cPLA(2) and iPLA(2) enzyme activities and cPLA(2) phosphorylation were stimulated after E. coli incubation and were attenuated by PLA(2), PI3-K, ERK 1/2 inhibitors. Our results demonstrate the role of PKCα/ERK/MAPK signaling pathways in governing the E. coli penetration into the brain.


Colloids and Surfaces B: Biointerfaces | 2017

Ferritin-supported lipid bilayers for triggering the endothelial cell response

Cristina Satriano; Gabriella Lupo; Carla Motta; Carmelina Daniela Anfuso; P. Di Pietro; Bengt Kasemo

Hybrid nanoassemblies of ferritin and silica-supported lipid bilayers (ferritin-SLBs) have been prepared and tested for the adhesion, spreading and proliferation of retinal microvascular endothelial cells (ECs). Lipid membranes with varying surface charge were obtained by mixing cationic 1-palmitoyl-2-oleoyl-sn-glycero-3-ethylphosphocholine (POEPC) with zwitterionic 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) at increasing POPC/POEPC ratios. The supported bilayer formation and their subsequent interaction processes with ferritin were studied at the pH of 7.4 at different protein concentrations, by using the quartz crystal microbalance with dissipation monitoring and by atomic force microscopy. Both kinetics and viscoelastic parameters of the protein-lipid membrane interface were scrutinized, as well as surface coverage. Phase-contrast optical microscopy analyses of the ferritin-SLBs substrates after their interaction with endothelial cells evidenced the highest cell adhesion (2-4h of incubation time) and proliferation (from 24h to 5 days) for the membranes of POPC/POEPC (75:25 ratio). Moreover, ferritin increased both cell adhesion and proliferation in comparison to control glass (respectively 1.5- and 1.75-fold) as well as proliferation in comparison to bare POPC/POEPC (95:5 ratio) (2 fold). Results are very promising in the goal of modulating the endothelial cell response through the interplay of viscoelastic/charge properties of the solid-supported membranes and the SLB-conditioned ferritin activity.

Collaboration


Dive into the Carla Motta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge