Network


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

Hotspot


Dive into the research topics where Valentina Pallottini is active.

Publication


Featured researches published by Valentina Pallottini.


Journal of Endocrinology | 2008

Sex differences in hepatic regulation of cholesterol homeostasis

Elisabetta De Marinis; Chiara Martini; Anna Trentalance; Valentina Pallottini

Physiological sex differences may influence metabolic status and then alter the onset of some diseases. According to recent studies, it is now well established that females are more protected from hypercholesterolemia-related diseases, such as cardiovascular diseases until menopause. Female protection from hypercholesterolemia is mediated by the hypolipidemic properties of estrogens, even if mechanisms underlying this protection remain still debated. Even though the regulatory mechanisms of cholesterol homeostasis maintenance are well known, few data are available on the supposed differences between male and female in these processes. So, the aim of this work was to define, through an in vivo study, the putative sex-dependent regulation of the processes underlying cholesterol homeostasis maintenance. We examined 3-hydroxy 3-methylglutaryl coenzyme A reductase and its regulatory protein network as well as the amount of low-density lipoprotein receptor and cholesterol. The study was conducted in the liver and plasma of male and female rats, on adults and during postnatal development, and on 17-beta-estradiol-treated male rats. Our data support that physiological differences in proteins involved in cholesterol balance are present between the sexes and, in particular, 3-hydroxy 3-methylglutaryl coenzyme A reductase shows lower activity and expression in female and 17-beta-estradiol-treated male rats than in adult untreated male. Our data suggest that sex differences in enzyme expression depend on variation in regulatory proteins and seem to be related to estrogen presence. This work adds new evidence in the complicated picture of sex-dependent cellular physiology and establishes a new role for reductase regulatory proteins as a link between estrogen protective effects and cholesterol homeostasis.


Molecular Nutrition & Food Research | 2009

Quercetin-induced apoptotic cascade in cancer cells: antioxidant versus estrogen receptor α-dependent mechanisms.

Paola Galluzzo; Chiara Martini; Pamela Bulzomi; Stefano Leone; Alessandro Bolli; Valentina Pallottini; Maria Marino

The flavonol quercetin, especially abundant in apple, wine, and onions, is reported to have anti-proliferative effects in many cancer cell lines. Antioxidant or pro-oxidant activities and kinase inhibition have been proposed as molecular mechanisms for these effects. In addition, an estrogenic activity has been observed but, at the present, it is poorly understood whether this latter activity plays a role in the quercetin-induced anti-proliferative effects. Here, we studied the molecular mechanisms of quercetin committed to the generation of an apoptotic cascade in cancer cells devoid or containing transfected estrogen receptor alpha (ERalpha; i.e., human cervix epitheloid carcinoma HeLa cells). Although none of tested quercetin concentrations increase reactive oxygen species (ROS) generation in HeLa cells, quercetin stimulation prevents the H(2)O(2)-induced ROS production both in the presence and in the absence of ERalpha. However, this flavonoid induces the activation of p38/MAPK, leading to the pro-apoptotic caspase-3 activation and to the poly(ADP-ribose) polymerase cleavage only in the presence of ERalpha. Notably, no down-regulation of survival kinases (i.e., AKT and ERK) was reported. Taken together, these findings suggest that quercetin results in HeLa cell death through an ERalpha-dependent mechanism involving caspase- and p38 kinase activation. These findings indicate new potential chemopreventive actions of flavonoids on cancer growth.


American Journal of Physiology-cell Physiology | 2009

17β-Estradiol regulates the first steps of skeletal muscle cell differentiation via ER-α-mediated signals

Paola Galluzzo; Chiara Rastelli; Pamela Bulzomi; Filippo Acconcia; Valentina Pallottini; Maria Marino

17beta-Estradiol (E(2)) mediates a wide variety of complex biological processes determining the growth and development of reproductive tract as well as nonreproductive tissues of male and female individuals. While E(2) effects on the reproductive system, bone, and cardiovascular system are quite well established, less is known about how it affects the physiology of other tissues. Skeletal muscle is a tissue that is expected to be E(2) responsive since both isoforms of estrogen receptor (ER-alpha and ER-beta) are expressed. Significant sex-related differences have been described in skeletal muscle, although the role played by E(2) and the mechanisms underlying it remain to be determined. Here, we demonstrate that E(2) increases the glucose transporter type 4 translocation at membranes as well as the expression of well-known differentiation markers of myogenesis (i.e., myogenin and myosin heavy chain) in rat myoblast cells (L6). These E(2)-induced effects require rapid extranuclear signals and the presence of ER-alpha, whereas no contribution of IGF-I receptor has been observed. In particular, ER-alpha-dependent Akt activation participates in regulating the first step of myogenic differentiation. Moreover, both receptors mediate the E(2)-induced activation of p38, which, in turn, affects the expression of myogenin and myosin heavy chain. All together, these data indicate that E(2) should be included in the list of skeletal muscle trophic factors.


Mechanisms of Ageing and Development | 2002

Age-related changes of cholesterol and dolichol biosynthesis in rat liver

Maria Marino; Valentina Pallottini; Cristina D'Eramo; Gabriella Cavallini; Ettore Bergamini; Anna Trentalance

Ageing has been defined as a gradually decreased ability to maintain homeostatic potential and increased risk to die, associated with a tissue accumulation of altered proteins and lipids. Among other, increased concentration of an isoprenoid compound, dolichol (Dol), in mammalian tissues during ageing has been reported and it has been considered as a new biomarker of ageing. However, the mechanism and the role of this accumulation is still unknown. Aim of this work was to study the mechanism of age-dependent Dol accumulation in the liver analysing the activity of the hepatic rate-limiting enzyme of isoprenoid biosynthesis, the 3-hydroxy 3-methylglutaryl CoA reductase (HMGCoA reductase), the Dol synthesis by mevalonate (MVA), the Dol level in the plasma, and the cholesterol (Chol) synthesis and content of ageing rat fed ad libitum (AL) or subjected to the effect of food restriction. Since the caloric restrictions are the most reproducible way to slow ageing and to extend life span, animals on these nutritional regimens were used to study ageing related mechanisms. The data show that during ageing the hepatic Dol accumulation is associated with an increase of HMGCoA reductase activity, which is affected by diet restriction, and with an increase of MVA incorporation in Dol and Chol, which is not. In addition, the liver of aged rats maintains the capability to regulate its Chol content and to modify Chol delivery into the blood.


World Journal of Hepatology | 2012

Regulation and deregulation of cholesterol homeostasis: The liver as a metabolic “power station”

Laura Trapani; Marco Segatto; Valentina Pallottini

Cholesterol plays several structural and metabolic roles that are vital for human biology. It spreads along the entire plasma membrane of the cell, modulating fluidity and concentrating in specialized sphingolipid-rich domains called rafts and caveolae. Cholesterol is also a substrate for steroid hormones. However, too much cholesterol can lead to pathological pictures such as atherosclerosis, which is a consequence of the accumulation of cholesterol into the cells of the artery wall. The liver is considered to be the metabolic power station of mammalians, where cholesterol homeostasis relies on an intricate network of cellular processes whose deregulations can lead to several life-threatening pathologies, such as familial and age-related hypercholesterolemia. Cholesterol homeostasis maintenance is carried out by: biosynthesis, via 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) activity; uptake, through low density lipoprotein receptors (LDLr); lipoprotein release in the blood; storage by esterification; and degradation and conversion into bile acids. Both HMGR and LDLr are transcribed as a function of cellular sterol amount by a family of transcription factors called sterol regulatory element binding proteins that are responsible for the maintenance of cholesterol homeostasis through an intricate mechanism of regulation. Cholesterol obtained by hepatic de novo synthesis can be esterified and incorporated into apolipoprotein B-100-containing very low density lipoproteins, which are then secreted into the bloodstream for transport to peripheral tissues. Moreover, dietary cholesterol is transferred from the intestine to the liver by high density lipoproteins (HDLs); all HDL particles are internalized in the liver, interacting with the hepatic scavenger receptor (SR-B1). Here we provide an updated overview of liver cholesterol metabolism regulation and deregulation and the causes of cholesterol metabolism-related diseases. Moreover, current pharmacological treatment and novel hypocholesterolemic strategies will also be introduced.


Journal of Cellular Physiology | 2001

β-estradiol stimulation of DNA synthesis requires different PKC isoforms in HEPG2 and MCF7 cells

Maria Marino; Enza Distefano; Simona Caporali; Gianna Ceracchi; Valentina Pallottini; Anna Trentalance

The role exerted by protein kinase C (PKC) on estrogen‐induced DNA synthesis has been investigated in hepatic and mammary gland cells, HepG2 and MCF7. 17‐β‐estradiol stimulated DNA synthesis in HepG2 and MCF7 cells, maximal effect occurring at 10 nM. DNA synthesis stimulation was prevented by anti‐estrogen ICI 182,780 and by inhibitor of PKC, Ro 31‐8220. The rapid estradiol effects in MCF7 cells were determined by following the inositol trisphosphate (IP3) production and PKC‐α membrane translocation. After estradiol treatment the increase of IP3 production, prevented by anti‐estrogen or by phospholipase C (PLC) inhibitor (neomycin), was present in MCF7 cells. In MDA cells, devoid of estrogen receptor, no effect was observed. The PKC‐α presence on the membranes appeared unchanged in MCF7 cells. The PLC inhibitors, neomycin and U73,122, and PKC‐α down regulator, phorbol 12‐myristate 13‐acetate (PMA), were able to prevent estradiol‐induced DNA synthesis in hepatoma cells, but ineffective in mammary cells; wortmannin, an inhibitor of phosphoinositide 3‐kinases (PI3‐K), blocked DNA synthesis in both cell lines. These data show that β‐estradiol, via an estrogen receptor‐mediated mechanism, activates more signal transduction pathways, and consequently different PKC isoforms in two responsive cell lines. In both cell lines PI3‐K/PKC pathway is functional to the estrogen regulation of DNA synthesis, whereas in HepG2 cells the parallel involvement of the PLC/PKC‐α pathway is present. The reported results indicate that the DNA synthesis stimulation by β‐estradiol requires the estrogen receptor and utilises one or more activated pathways in dependence on the cell equipment.


Mechanisms of Ageing and Development | 2007

Age-related HMG-CoA reductase deregulation depends on ROS-induced p38 activation.

Valentina Pallottini; Chiara Martini; Gabriella Cavallini; Ettore Bergamini; Kirsty J. Mustard; D. Grahame Hardie; Anna Trentalance

BACKGROUND It seems to be clear that hepatic age-related HMG-CoA reductase total activation is connected to a rise of reactive oxygen species (ROS). However, the mechanism by which ROS achieve this effect is unknown. Thus, in this work, we have performed a study of HMG-CoAR by analyzing the enzymes involved in its short-term regulation, namely, AMP-activated kinase (AMPK) and protein phosphatase 2A (PP2A). METHODS AND MATERIALS In the liver of aged rats and in H(2)O(2)-stimulated HepG2 cells the ROS content, the HMG-CoA reductase activation state, its regulatory enzymes and the p38 downstream pathway involved in reductase deregulation, have been studied. RESULTS AND CONCLUSIONS Our data show that the hepatic HMG-CoAR is completely dephosphorylated in the liver of old rat being the PP2A increased association with HMG-CoAR the main responsible. On the other hand, the age-related greater association between PP2A and HMG-CoAR results to be due to an increase in ROS that is present during aging and has already been demonstrated to influence HMG-CoAR activation state. Moreover, H(2)O(2)-stimulated HepG2 cell line shows that the ROS effect on the HMG-CoAR dephosphorylation is mediated by the activation of p38/MAPK pathway.


Iubmb Life | 2011

Potential role of nonstatin cholesterol lowering agents

Laura Trapani; Marco Segatto; Paolo Ascenzi; Valentina Pallottini

Although statins, 3β‐hydroxy‐3β‐methylglutaryl coenzyme A reductase (HMGR) inhibitors, have revolutionized the management of cardiovascular diseases by lowering serum low density lipoproteins, many patients suffer from their side effects. Whether the statin side effects are related to their intrinsic toxicity or to the decrease of HMGR main isoprenoid end products, which are essential compounds for cell viability, is still debated. In addition to HMGR, the key and rate limiting step of cholesterol synthesis, many enzymes are involved in this multi‐step pathway whose inhibition could be taken into account for a “nonstatin approach” in the management of hypercholesterolemia. In particular, due to their unique position downstream from HMGR, the inhibition of squalene synthase, farnesyl diphosphate farnesyltransferase (FDFT1), squalene epoxidase (SQLE), and oxidosqualene cyclase:lanosterol synthase (OSC) should decrease plasma levels of cholesterol without affecting ubiquinone, dolichol, and isoprenoid metabolism. Thus, although FDFT1, SQLE and OSC are little studied, they should be considered as perspective targets for the development of novel drugs against hypercholesterolemia. Here, structure–function relationships of FDFT1, SQLE, and OSC are reviewed highlighting the advantages that the downstream inhibition of HMGR could provide when compared to the statin‐based therapy.


Biochimica et Biophysica Acta | 2002

Role of tyrosine kinase signaling in estrogen-induced LDL receptor gene expression in HepG2 cells

Enza Distefano; Maria Marino; Jennifer Gillette; Bettina Hanstein; Valentina Pallottini; Jens C. Brüning; Wilhelm Krone; Anna Trentalance

The expression of the low-density lipoprotein receptor (LDL-r) gene is stimulated by estrogen in vivo, although its promoter does not contain a classical estrogen-responsive element, suggesting an alternative mechanism of estrogen-regulated expression of this gene. The aim of this work was to assess whether estrogen-stimulated transcription of the LDL-r gene depends on tyrosine kinase (TK) and protein kinase C (PKC) activation, both signaling pathways being activated by estrogen in vivo and in hepatoma cells. Therefore, in HepG2 cells cotransfected with estrogen receptor-alpha, estrogen-stimulated transcription of LDL-r-promoter reporter plasmid was analyzed in the absence and presence of TK and PKC inhibitors. The expression of LDL-r was also compared with the transcription of the complement gene, which contains a classical estrogen-responsive element sequence. Our results demonstrate that the induction of LDL-r expression by estrogen requires longer stimulation than that necessary for complement induction. Moreover, basal transcription of the LDL-r gene depends on PKC activity, while estrogen-stimulated activation of the LDL-r-promoter requires TK activity, pointing to a role of these non-classical estrogen-stimulated pathways in the transcriptional regulation of the LDL-r.


Journal of Cellular Biochemistry | 2006

Modified HMG-CoA reductase and LDLr regulation is deeply involved in age-related hypercholesterolemia.

Valentina Pallottini; Chiara Martini; Gabriella Cavallini; Alessio Donati; Ettore Bergamini; Maria Notarnicola; Maria Gabriella Caruso; Anna Trentalance

During the ageing process in rats hypercholesterolemia occurs in concert with full activation, lowered degradation rate and an unchanged level of the rate limiting cholesterol biosynthesis enzyme, 3‐hydroxy‐3‐methylglutaryl coenzyme A reductase (HMG‐CoAR). The molecular bases of the HMG‐CoAR unchanged level and lowered degradation rate in aged rats is not clear. In fact no data are available during ageing, on transcription and degradation of HMG‐CoAR, so well defined in adult animal. So, aim of this work was to measure mRNA levels of the enzyme and the level of the proteins of the regulatory complex responsible of the cholesterol metabolism. To complete the picture, the level of sterol regulatory element binding proteins (SREBPs), SREBP cleavage activating protein, and insulin‐induced gene has been measured. The levels of other related proteins, whose transcription is SREBP dependent, that is low density lipoprotein receptor (LDLr) and Caveolin 1, have been also measured. The age‐related reduced Insigs levels, joined to a reduced insulin sensitivity, could explain the decreased degradation rate of the HMG‐CoAR and the increased active SREBP‐2. The SREBP‐2 in particular seems to be committed in multiple way to gene transcription. The obtained data represent a good contribution to explain the age‐related hypercholesterolemia. J. Cell. Biochem. 98: 1044–1053, 2006.

Collaboration


Dive into the Valentina Pallottini's collaboration.

Top Co-Authors

Avatar

Anna Trentalance

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chiara Martini

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge