22 results on '"Grazia Scandura"'
Search Results
2. PB2093: OVERALL SURVIVAL IN RELAPSED-REFRACTORY MYELOMA PATIENTS DEPENDS ON CIRCULATING PLASMA CELLS QUANTITATION
- Author
-
Nunziatina Laura Parrinello, Grazia Scandura, Sara Marino, Anna Triolo, Vittorio Del Fabro, Francesco DI Raimondo, Concetta Conticello, and Alessandra Romano
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. P852: ARGININE DEPRIVATION INDUCES ACQUISITION OF A SENESCENT PHENOTYPE AND FAVORS GENOMIC INSTABILITY IN MULTIPLE MYELOMA PLASMA CELLS
- Author
-
Alessandra Romano, Grazia Scandura, Cesarina Giallongo, Enrico La Spina, Lucia Longhitano, Tatiana Zuppelli, Ilaria Dulcamare, Nunziatina Laura Parrinello, Francesca Polito, Rosaria Oteri, Mohammed Aguennouz, Nunzio Vicario, Angela Maria Amorini, Vittorio Del Fabro, Concetta Conticello, Giovanni LI Volti, Giuseppe Alberto Palumbo, Daniele Tibullo, and Francesco DI Raimondo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
4. Engagement of Mesenchymal Stromal Cells in the Remodeling of the Bone Marrow Microenvironment in Hematological Cancers
- Author
-
Sebastiano Giallongo, Andrea Duminuco, Ilaria Dulcamare, Tatiana Zuppelli, Enrico La Spina, Grazia Scandura, Annalisa Santisi, Alessandra Romano, Francesco Di Raimondo, Daniele Tibullo, Giuseppe A. Palumbo, and Cesarina Giallongo
- Subjects
MSCs ,tumor transformation ,hematological cancers ,senescence ,inflammation ,Microbiology ,QR1-502 - Abstract
Mesenchymal stromal cells (MSCs) are a subset of heterogeneous, non-hematopoietic fibroblast-like cells which play important roles in tissue repair, inflammation, and immune modulation. MSCs residing in the bone marrow microenvironment (BMME) functionally interact with hematopoietic stem progenitor cells regulating hematopoiesis. However, MSCs have also emerged in recent years as key regulators of the tumor microenvironment. Indeed, they are now considered active players in the pathophysiology of hematologic malignancies rather than passive bystanders in the hematopoietic microenvironment. Once a malignant event occurs, the BMME acquires cellular, molecular, and epigenetic abnormalities affecting tumor growth and progression. In this context, MSC behavior is affected by signals coming from cancer cells. Furthermore, it has been shown that stromal cells themselves play a major role in several hematological malignancies’ pathogenesis. This bidirectional crosstalk creates a functional tumor niche unit wherein tumor cells acquire a selective advantage over their normal counterparts and are protected from drug treatment. It is therefore of critical importance to unveil the underlying mechanisms which activate a protumor phenotype of MSCs for defining the unmasked vulnerabilities of hematological cancer cells which could be pharmacologically exploited to disrupt tumor/MSC coupling. The present review focuses on the current knowledge about MSC dysfunction mechanisms in the BMME of hematological cancers, sustaining tumor growth, immune escape, and cancer progression.
- Published
- 2023
- Full Text
- View/download PDF
5. CXCL12/CXCR4 axis supports mitochondrial trafficking in tumor myeloma microenvironment
- Author
-
Cesarina Giallongo, Ilaria Dulcamare, Daniele Tibullo, Vittorio Del Fabro, Nunzio Vicario, Nunziatina Parrinello, Alessandra Romano, Grazia Scandura, Giacomo Lazzarino, Concetta Conticello, Giovanni Li Volti, Angela Maria Amorini, Giuseppe Musumeci, Michelino Di Rosa, Francesca Polito, Rosaria Oteri, M’hammed Aguennouz, Rosalba Parenti, Francesco Di Raimondo, and Giuseppe A. Palumbo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Mesenchymal stromal cells (MSCs) within the protective microenvironment of multiple myeloma (MM) promote tumor growth, confer chemoresistance and support metabolic needs of plasma cells (PCs) even transferring mitochondria. In this scenario, heterocellular communication and dysregulation of critical signaling axes are among the major contributors to progression and treatment failure. Here, we report that myeloma MSCs have decreased reliance on mitochondrial metabolism as compared to healthy MSCs and increased tendency to deliver mitochondria to MM cells, suggesting that this intercellular exchange between PCs and stromal cells can be consider part of MSC pro-tumorigenic phenotype. Interestingly, we also showed that PCs promoted expression of connexin 43 (CX43) in MSCs leading to CXCL12 activation and stimulation of its receptor CXCR4 on MM cells favoring protumor mitochondrial transfer. Consistently, we observed that selective inhibition of CXCR4 by plerixafor resulted in a significant reduction of mitochondria trafficking. Moreover, intracellular expression of CXCR4 in myeloma PCs from BM biopsy specimens demonstrated higher CXCR4 colocalization with CD138+ cells of non-responder patients to bortezomib compared with responder patients, suggesting that CXCR4 mediated chemoresistance in MM. Taken together, our data demonstrated that CXCL12/CXCR4 axis mediates intercellular coupling thus suggesting that the myeloma niche may be exploited as a target to improve and develop therapeutic approaches.
- Published
- 2022
- Full Text
- View/download PDF
6. Impact of buffer composition on biochemical, morphological and mechanical parameters: A tare before dielectrophoretic cell separation and isolation
- Author
-
Paolo G. Bonacci, Giuseppe Caruso, Grazia Scandura, Clarissa Pandino, Alessandra Romano, Giorgio I. Russo, Ronald Pethig, Massimo Camarda, and Nicolò Musso
- Subjects
Dielectrophoresis ,Cell status ,Biochemical parameters ,Circulating tumor cells ,Cell isolation and separation ,Microfluidics ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Dielectrophoresis (DEP) represents an electrokinetic approach for discriminating and separating suspended cells based on their intrinsic dielectric characteristics without the need for labeling procedure. A good practice, beyond the physical and engineering components, is the selection of a buffer that does not hinder cellular and biochemical parameters as well as cell recovery. In the present work the impact of four buffers on biochemical, morphological, and mechanical parameters was evaluated in two different cancer cell lines (Caco-2 and K562). Specifically, MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide]) assay along with flow cytometry analysis were used to evaluate the occurring changes in terms of cell viability, morphology, and granulocyte stress formation, all factors directly influencing DEP sorting capability. Quantitative real-time PCR (qRT-PCR) was instead employed to evaluate the gene expression levels of interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS), two well-known markers of inflammation and oxidative stress, respectively. An additional marker representing an index of cellular metabolic status, i.e. the expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene, was also evaluated. Among the four buffers considered, two resulted satisfactory in terms of cell viability and growth recovery (24 h), with no significant changes in cell morphology for up to 1 h in suspension. Of note, gene expression analysis showed that in both cell lines the apparently non-cytotoxic buffers significantly modulated IL-6, iNOS, and GAPDH markers, underlining the importance to deeply investigate the molecular and biochemical changes occurring during the analysis, even at apparently non-toxic conditions. The selection of a useful buffer for the separation and analysis of cells without labeling procedures, preserving cell status, represents a key factor for DEP analysis, giving the opportunity to further use cells for additional analysis.
- Published
- 2023
- Full Text
- View/download PDF
7. Clobetasol promotes neuromuscular plasticity in mice after motoneuronal loss via sonic hedgehog signaling, immunomodulation and metabolic rebalancing
- Author
-
Nunzio Vicario, Federica M. Spitale, Daniele Tibullo, Cesarina Giallongo, Angela M. Amorini, Grazia Scandura, Graziana Spoto, Miriam W. Saab, Simona D’Aprile, Cristiana Alberghina, Renata Mangione, Joshua D. Bernstock, Cirino Botta, Massimo Gulisano, Emanuele Buratti, Giampiero Leanza, Robert Zorec, Michele Vecchio, Michelino Di Rosa, Giovanni Li Volti, Giuseppe Lazzarino, Rosalba Parenti, and Rosario Gulino
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Motoneuronal loss is the main feature of amyotrophic lateral sclerosis, although pathogenesis is extremely complex involving both neural and muscle cells. In order to translationally engage the sonic hedgehog pathway, which is a promising target for neural regeneration, recent studies have reported on the neuroprotective effects of clobetasol, an FDA-approved glucocorticoid, able to activate this pathway via smoothened. Herein we sought to examine functional, cellular, and metabolic effects of clobetasol in a neurotoxic mouse model of spinal motoneuronal loss. We found that clobetasol reduces muscle denervation and motor impairments in part by restoring sonic hedgehog signaling and supporting spinal plasticity. These effects were coupled with reduced pro-inflammatory microglia and reactive astrogliosis, reduced muscle atrophy, and support of mitochondrial integrity and metabolism. Our results suggest that clobetasol stimulates a series of compensatory processes and therefore represents a translational approach for intractable denervating and neurodegenerative disorders.
- Published
- 2021
- Full Text
- View/download PDF
8. P042: Metabolic regulation of adaptive response to arginine deprivation in Hodgkin Lymphoma
- Author
-
Alessandra Romano, Grazia Scandura, Gabriella Santuccio, Fabrizio Puglisi, Daniela Cambria, Angela Maria Amorini, Enrico La Spina, Nunziatina Laura Parrinello, Cesarina Giallongo, Daniele Tibullo, Claudio Cerchione, Giovanni Martinelli, Giorgia Simonetti, and Francesco Di Raimondo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
9. Mechanisms of Action of the New Antibodies in Use in Multiple Myeloma
- Author
-
Alessandra Romano, Paola Storti, Valentina Marchica, Grazia Scandura, Laura Notarfranchi, Luisa Craviotto, Francesco Di Raimondo, and Nicola Giuliani
- Subjects
monoclonal antibodies ,multiple myeloma ,CD38 ,SLAMF7 ,BCMA ,antibody-drug conjugate ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Monoclonal antibodies (mAbs) directed against antigen-specific of multiple myeloma (MM) cells have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP), but the choice of the antigen is crucial for the development of effective immuno-therapy in MM. Recently new immunotherapeutic options in MM patients have been developed against different myeloma-related antigens as drug conjugate-antibody, bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR)-T cells. In this review, we will highlight the mechanism of action of immuno-therapy currently available in clinical practice to target CD38, SLAMF7, and BCMA, focusing on the biological role of the targets and on mechanisms of actions of the different immunotherapeutic approaches underlying their advantages and disadvantages with critical review of the literature data.
- Published
- 2021
- Full Text
- View/download PDF
10. TLR4 Signaling and Heme Oxygenase-1/Carbon Monoxide Pathway Crosstalk Induces Resiliency of Myeloma Plasma Cells to Bortezomib Treatment
- Author
-
Grazia Scandura, Cesarina Giallongo, Fabrizio Puglisi, Alessandra Romano, Nunziatina Laura Parrinello, Tatiana Zuppelli, Lucia Longhitano, Sebastiano Giallongo, Michelino Di Rosa, Giuseppe Musumeci, Roberto Motterlini, Roberta Foresti, Giuseppe Alberto Palumbo, Giovanni Li Volti, Francesco Di Raimondo, and Daniele Tibullo
- Subjects
multiple myeloma ,TLR4/HO-1 crosstalk ,mitochondria ,bortezomib ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Relapse in multiple myeloma (MM) decreases therapy efficiency through unclear mechanisms of chemoresistance. Since our group previously demonstrated that heme oxygenase-1 (HO-1) and Toll-like receptor 4 (TLR4) are two signaling pathways protecting MM cells from the proteasome inhibitor bortezomib (BTZ), we here evaluated their cross-regulation by a pharmacological approach. We found that cell toxicity and mitochondrial depolarization by BTZ were increased upon inhibition of HO-1 and TLR4 by using tin protoporphyrin IX (SnPP) and TAK-242, respectively. Furthermore, the combination of TAK-242 and BTZ activated mitophagy and decreased the unfolded protein response (UPR) survival pathway in association with a downregulation in HO-1 expression. Notably, BTZ in combination with SnPP induced effects mirroring the treatment with TAK-242/BTZ, resulting in a blockade of TLR4 upregulation. Interestingly, treatment of cells with either hemin, an HO-1 inducer, or supplementation with carbon monoxide (CO), a by-product of HO-1 enzymatic activity, increased TLR4 expression. In conclusion, we showed that treatment of MM cells with BTZ triggers the TLR4/HO-1/CO axis, serving as a stress-responsive signal that leads to increased cell survival while protecting mitochondria against BTZ and ultimately promoting drug resistance.
- Published
- 2022
- Full Text
- View/download PDF
11. Mitochondrial Bioenergetics at the Onset of Drug Resistance in Hematological Malignancies: An Overview
- Author
-
Alessandro Barbato, Grazia Scandura, Fabrizio Puglisi, Daniela Cambria, Enrico La Spina, Giuseppe Alberto Palumbo, Giacomo Lazzarino, Daniele Tibullo, Francesco Di Raimondo, Cesarina Giallongo, and Alessandra Romano
- Subjects
OX-PHOS ,mitochondria ,multiple myeloma ,acute myeloid leukemia ,chronic lymphatic leukemia ,lymphoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The combined derangements in mitochondria network, function and dynamics can affect metabolism and ATP production, redox homeostasis and apoptosis triggering, contributing to cancer development in many different complex ways. In hematological malignancies, there is a strong relationship between cellular metabolism, mitochondrial bioenergetics, interconnections with supportive microenvironment and drug resistance. Lymphoma and chronic lymphocytic leukemia cells, e.g., adapt to intrinsic oxidative stress by increasing mitochondrial biogenesis. In other hematological disorders such as myeloma, on the contrary, bioenergetics changes, associated to increased mitochondrial fitness, derive from the adaptive response to drug-induced stress. In the bone marrow niche, a reverse Warburg effect has been recently described, consisting in metabolic changes occurring in stromal cells in the attempt to metabolically support adjacent cancer cells. Moreover, a physiological dynamic, based on mitochondria transfer, between tumor cells and their supporting stromal microenvironment has been described to sustain oxidative stress associated to proteostasis maintenance in multiple myeloma and leukemia. Increased mitochondrial biogenesis of tumor cells associated to acquisition of new mitochondria transferred by mesenchymal stromal cells results in augmented ATP production through increased oxidative phosphorylation (OX-PHOS), higher drug resistance, and resurgence after treatment. Accordingly, targeting mitochondrial biogenesis, electron transfer, mitochondrial DNA replication, or mitochondrial fatty acid transport increases therapy efficacy. In this review, we summarize selected examples of the mitochondrial derangements in hematological malignancies, which provide metabolic adaptation and apoptosis resistance, also supported by the crosstalk with tumor microenvironment. This field promises a rational design to improve target-therapy including the metabolic phenotype.
- Published
- 2020
- Full Text
- View/download PDF
12. Focus on Osteosclerotic Progression in Primary Myelofibrosis
- Author
-
Mariarita Spampinato, Cesarina Giallongo, Alessandra Romano, Lucia Longhitano, Enrico La Spina, Roberto Avola, Grazia Scandura, Ilaria Dulcamare, Vincenzo Bramanti, Michelino Di Rosa, Nunzio Vicario, Rosalba Parenti, Giovanni Li Volti, Daniele Tibullo, and Giuseppe A. Palumbo
- Subjects
primary myelofibrosis ,bone ,myeloproliferative neoplasm ,bone marrow ,fibrosis ,Microbiology ,QR1-502 - Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
- Published
- 2021
- Full Text
- View/download PDF
13. Lactate trafficking inhibition restores sensitivity to proteasome inhibitors and orchestrates immuno-microenvironment in multiple myeloma
- Author
-
Alessandro Barbato, Cesarina Giallongo, Sebastiano Giallongo, Alessandra Romano, Grazia Scandura, Saoca Concetta, Tatiana Zuppelli, Marco Lolicato, Giacomo Lazzarino, Nunziatina Parrinello, Vittorio Del Fabro, Paolo Fontana, M'hammed Aguennoz, Giovanni Li Volti, Giuseppe A. Palumbo, Francesco Di Raimondo, and Daniele Tibullo
- Subjects
Cell Biology ,General Medicine - Published
- 2023
14. Impact of buffer composition on biochemical, morphological and mechanical parameters: A tare before dielectrophoretic cell separation and isolation
- Author
-
Paolo G. Bonacci, Giuseppe Caruso, Grazia Scandura, Clarissa Pandino, Alessandra Romano, Giorgio I. Russo, Ronald Pethig, Massimo Camarda, and Nicolò Musso
- Subjects
Cancer Research ,Oncology - Abstract
Dielectrophoresis (DEP) represents an electrokinetic approach for discriminating and separating suspended cells based on their intrinsic dielectric characteristics without the need for labeling procedure. A good practice, beyond the physical and engineering components, is the selection of a buffer that does not hinder cellular and biochemical parameters as well as cell recovery. In the present work the impact of four buffers on biochemical, morphological, and mechanical parameters was evaluated in two different cancer cell lines (Caco-2 and K562). Specifically, MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide]) assay along with flow cytometry analysis were used to evaluate the occurring changes in terms of cell viability, morphology, and granulocyte stress formation, all factors directly influencing DEP sorting capability. Quantitative real-time PCR (qRT-PCR) was instead employed to evaluate the gene expression levels of interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS), two well-known markers of inflammation and oxidative stress, respectively. An additional marker representing an index of cellular metabolic status, i.e. the expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene, was also evaluated. Among the four buffers considered, two resulted satisfactory in terms of cell viability and growth recovery (24 h), with no significant changes in cell morphology for up to 1 h in suspension. Of note, gene expression analysis showed that in both cell lines the apparently non-cytotoxic buffers significantly modulated IL-6, iNOS, and GAPDH markers, underlining the importance to deeply investigate the molecular and biochemical changes occurring during the analysis, even at apparently non-toxic conditions. The selection of a useful buffer for the separation and analysis of cells without labeling procedures, preserving cell status, represents a key factor for DEP analysis, giving the opportunity to further use cells for additional analysis.
- Published
- 2022
15. Arginine Deprivation in Microenvironment Makes Multiple Myeloma Resistant to Proteasome Inhibitors Via Increased Lactate Release and Metabolic Rewiring
- Author
-
Cesarina Giallongo, Grazia Scandura, Alessandro Barbato, Enrico La Spina, Lucia Longhitano, Tatiana Zuppelli, Ilaria Dulcamare, Nunziatina Laura Parrinello, Sebastiano Giallongo, Francesca Polito, Rosaria Oteri, Mohammed Aguennouz, Giuseppe A. Palumbo, Marco Lolicato, Giovanni Li Volti, Francesco Di Raimondo, Daniele Tibullo, and Alessandra Romano
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
16. Clobetasol promotes neuromuscular plasticity in mice after motoneuronal loss via sonic hedgehog signaling, immunomodulation and metabolic rebalancing
- Author
-
Graziana Spoto, Michele Vecchio, Angela Maria Amorini, Michelino Di Rosa, Rosalba Parenti, Rosario Gulino, Nunzio Vicario, Grazia Scandura, Federica M. Spitale, Cirino Botta, Simona D’Aprile, G. Leanza, Massimo Gulisano, Giuseppe Lazzarino, Joshua D. Bernstock, Emanuele Buratti, Daniele Tibullo, Cristiana Alberghina, Cesarina Giallongo, Renata Mangione, Miriam Wissam Saab, Robert Zorec, Giovanni Li Volti, Vicario N., Spitale F.M., Tibullo D., Giallongo C., Amorini A.M., Scandura G., Spoto G., Saab M.W., D'Aprile S., Alberghina C., Mangione R., Bernstock J.D., Botta C., Gulisano M., Buratti E., Leanza G., Zorec R., Vecchio M., Di Rosa M., Li Volti G., Lazzarino G., Parenti R., and Gulino R.
- Subjects
Male ,Cancer Research ,Physiology ,129 Strain ,Biochemistry ,Mice ,Databases, Genetic ,Medicine ,Myocyte ,Motor Neurons ,Neuronal Plasticity ,Skeletal ,Smoothened Receptor ,Hedgehog signaling pathway ,Muscle atrophy ,Mitochondria ,Astrogliosis ,Neuroprotective Agents ,Muscle ,medicine.symptom ,Inflammation Mediators ,Signal Transduction ,Cholera Toxin ,Mice, 129 Strain ,hedgehog ,Immunology ,Motor Activity ,Neuroprotection ,Article ,Databases ,Cellular and Molecular Neuroscience ,smoothened ,Genetic ,Animals ,Humans ,Hedgehog Proteins ,Muscle, Skeletal ,Hedgehog ,Glucocorticoids ,Muscle Denervation ,QH573-671 ,Animal ,business.industry ,Amyotrophic Lateral Sclerosis ,Glial biology ,Cell Biology ,medicine.disease ,Saporins ,Spine ,Mitochondria, Muscle ,Disease Models, Animal ,clobetasol ,inflammation ,Case-Control Studies ,Disease Models ,Diseases of the nervous system ,Cytology ,Smoothened ,business ,Energy Metabolism ,Neuroscience ,Open Field Test - Abstract
Motoneuronal loss is the main feature of amyotrophic lateral sclerosis, although pathogenesis is extremely complex involving both neural and muscle cells. In order to translationally engage the sonic hedgehog pathway, which is a promising target for neural regeneration, recent studies have reported on the neuroprotective effects of clobetasol, an FDA-approved glucocorticoid, able to activate this pathway via smoothened. Herein we sought to examine functional, cellular, and metabolic effects of clobetasol in a neurotoxic mouse model of spinal motoneuronal loss. We found that clobetasol reduces muscle denervation and motor impairments in part by restoring sonic hedgehog signaling and supporting spinal plasticity. These effects were coupled with reduced pro-inflammatory microglia and reactive astrogliosis, reduced muscle atrophy, and support of mitochondrial integrity and metabolism. Our results suggest that clobetasol stimulates a series of compensatory processes and therefore represents a translational approach for intractable denervating and neurodegenerative disorders.
- Published
- 2021
17. Mitochondrial Bioenergetics at the Onset of Drug Resistance in Hematological Malignancies: An Overview
- Author
-
Cesarina Giallongo, Grazia Scandura, Giuseppe A. Palumbo, Daniela Cambria, Alessandra Romano, Francesco Di Raimondo, Giacomo Lazzarino, Alessandro Barbato, Fabrizio Puglisi, Enrico La Spina, and Daniele Tibullo
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,lymphoma ,Oxidative phosphorylation ,Review ,Mitochondrion ,Biology ,acute myeloid leukemia ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Cell biology ,mitochondria ,multiple myeloma ,Proteostasis ,Mitochondrial biogenesis ,Oncology ,Reverse Warburg effect ,chronic lymphatic leukemia ,Mitochondrial DNA replication ,OX-PHOS - Abstract
The combined derangements in mitochondria network, function and dynamics can affect metabolism and ATP production, redox homeostasis and apoptosis triggering, contributing to cancer development in many different complex ways. In hematological malignancies, there is a strong relationship between cellular metabolism, mitochondrial bioenergetics, interconnections with supportive microenvironment and drug resistance. Lymphoma and chronic lymphocytic leukemia cells, e.g., adapt to intrinsic oxidative stress by increasing mitochondrial biogenesis. In other hematological disorders such as myeloma, on the contrary, bioenergetics changes, associated to increased mitochondrial fitness, derive from the adaptive response to drug-induced stress. In the bone marrow niche, a reverse Warburg effect has been recently described, consisting in metabolic changes occurring in stromal cells in the attempt to metabolically support adjacent cancer cells. Moreover, a physiological dynamic, based on mitochondria transfer, between tumor cells and their supporting stromal microenvironment has been described to sustain oxidative stress associated to proteostasis maintenance in multiple myeloma and leukemia. Increased mitochondrial biogenesis of tumor cells associated to acquisition of new mitochondria transferred by mesenchymal stromal cells results in augmented ATP production through increased oxidative phosphorylation (OX-PHOS), higher drug resistance, and resurgence after treatment. Accordingly, targeting mitochondrial biogenesis, electron transfer, mitochondrial DNA replication, or mitochondrial fatty acid transport increases therapy efficacy. In this review, we summarize selected examples of the mitochondrial derangements in hematological malignancies, which provide metabolic adaptation and apoptosis resistance, also supported by the crosstalk with tumor microenvironment. This field promises a rational design to improve target-therapy including the metabolic phenotype.
- Published
- 2020
18. Dissecting the Adaptive Response to Arginine Deprivation in Hodgkin Lymphoma
- Author
-
Francesco Di Raimondo, Grazia Scandura, Daniela Cambria, Antonella Padella, Fabrizio Puglisi, Alessandro Barbato, Giorgia Simonetti, Claudio Cerchione, Daniele Tibullo, Giovanni Martinelli, Enrico La Spina, Cesarina Giallongo, Nunziatina Laura Parrinello, Alessandra Romano, and Giuseppe A. Palumbo
- Subjects
Arginine deprivation ,business.industry ,Immunology ,Cancer research ,Hodgkin lymphoma ,Medicine ,Cell Biology ,Hematology ,Adaptive response ,business ,Biochemistry - Abstract
Background In Hodgkin Lymphoma (HL), neoplastic cells orchestrate an inflammatory microenvironment leading to sterile inflammation, T-cell anergy, and immune deficiency. Our group showed that in HL patients the aminoacid degrading enzyme Arginase-1 is increased, associated with poor outcome, and leads to arginine (Arg) deprivation. However, how the reduction of Arg in the extracellular milieu of the tumor microenvironment can contribute to neoplastic cell fitness is largely unknown. Aims To detect the adaptive response (via evaluation of global transcriptome and metabolome changes) in human HL cell lines exposed to Arg deprivation. Methods To better understand the impact of extra-cellular Arg1 deprivation on the metabolome of human cHL cells, four human cHL cell lines (L428, L540, HDMYZ and KM-H2) were individually cultured with customized complete media or lacking or Arg (R0), supplemented with 10% dialyzed fetal bovine serum, in six independent experiments. After 48 hours of culture, the cells were collected for global metabolomic analysis, by gas chromatography-mass spectrometry (GC/MS) and liquid chromatography-tandem mass spectrometry (LC/MS/MS) platforms by Metabolon Inc and transcriptome profiling by Illumina platform. Following normalization to DNA concentration, log transformation, and imputation of missing values, if any, with the minimum observed value for each compound, Welch's two-sample t-test was used to identify biochemicals that differed significantly between experimental groups. Results While Arg deprivation did not affect cell viability but delayed cell cycle due to arrest in G2 phase in all tested cell lines, the effect of Arg deficiency on the cellular metabolome depended largely on the cell type examined with L428 and KMH2 cells having significantly changed metabolomes. Pyruvate was significantly higher in the KMH2 cells deprived of Arg compared to controls. Conversely, lactate was significantly lower, with increased levels of long-chain saturated fatty acids and long-chain polyunsaturated fatty acids (PUFAs). Taken together the metabolomics changes suggested that specific-amino acid deficiency can lead to an increase in free fatty acids synthases to preserve cytoplasmatic and mitochondrial membrane dynamics. Consistent with a metabolic rewiring to maintain mitochondrial integrity (the pyruvate is an important intermediary in the conversion of carbohydrates into fatty acids), the adaptive response was associated to increased oxidative stress, as suggested by of reduced glutathione in KMH2 cells, depletion of gamma-glutamylcysteine, increased cystine, the oxidative product of cysteine, and methionine sulfoxide (an oxidation product of methionine). Gene set enrichment analysis (GSEA) showed deep transcriptome rearrangements in KMH2 and HDMYZ cell lines, involving upregulation of genes required for the unfolded protein response (UPR, including XBP1, EIF2S1, EIF4A2, EIF4A3, ATF3, ATF4, DDIT4, EDEM1, GADD45B, SQSTM1, HMOX), NF-kB response to TNF (including RAF1, TNF, LIF, NKBIA, SGK1, BIRC3, ICAM1, BCL6, IL6, RELA, CDKN1A), p53 pathway and networks (including CDKN2B, STOM, TRAF4, RRAD, SESN1, FOXO3, SERPINB5, JAG2) and proteosome degradation (HSPA4, PSMD11, PSMD13, PSMD2, PSMA5, PSMA7, PSMC4), with a minimal effect on metabolism features, except the upregulation of genes involved in lactate generation and degradation. All lines tested showed down-regulation of CCNI2, LCROL, MKI67, NCAPG, PEX10 and UFSP2, suggesting that early response to arginine deprivation includes modulation of UFMylation pathway, the most recently discovered post-translational protein modification system, whose biological function is largely unknown. Conclusions The removal of Arg from L428 and KMH2 resulted in changes in the specific-amino acid-related metabolites. The adaptive response to Arg-depleted environment increases oxidative stress and promotes a shift in glucose use in the attempt to preserve mitochondrial function. The cell-cycle arrest in G2, the increase of pyruvate availability and the upregulation of proteasome function via upregulation of the UFMylation pathway suggest the dependency of HL cell lines on mitochondrial function integrity. Quantity and function of mitochondria network can play a major role in selecting the fittest clones, a metabolic pathway that should be explored as novel non -synthetic lethal targets. Disclosures Martinelli: Stemline Therapeutics: Consultancy; Roche: Consultancy; Astellas: Consultancy, Speakers Bureau; Daichii Sankyo: Consultancy; Pfizer: Consultancy, Speakers Bureau; Incyte: Consultancy; Abbvie: Consultancy; Celgene /BMS: Consultancy, Speakers Bureau; Jazz Pharmaceuticals: Consultancy. Di Raimondo: Pfizer: Honoraria; Jazz Pharmaceutical: Honoraria; Amgen: Honoraria; AbbVie: Honoraria; Bristol Myers Squibb: Honoraria; Janssen Pharmaceuticals: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
19. CXCL12/CXCR4 Axis Drives Mitochondrial Trafficking in Tumor Myeloma Microenvironment
- Author
-
Grazia Scandura, Enrico La Spina, Ilaria Dulcamare, Giuseppe Musumeci, Alessandro Barbato, Giovanni Li Volti, Alessandra Romano, Giuseppe A. Palumbo, Concetta Conticello, Daniela Cambria, Nunzio Vicario, Vittorio Del Fabro, Nunziatina Laura Parrinello, Cesarina Giallongo, Giacomo Lazzarino, Daniele Tibullo, and Francesco Di Raimondo
- Subjects
Immunology ,Cancer research ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 - Abstract
Mesenchymal stromal cells (MSCs) within the protective microenvironment of multiple myeloma (MM) promote tumor growth, confer chemoresistance and support metabolic needs of plasma cells (PCs) also transferring mitochondria. In this scenario, heterocellular communication and dysregulation of critical signaling axes are among the major contributors to progression and treatment failure. As metabolic rewiring is involved in the regulation of MSC phenotype, we first analyzed metabolic profile of healthy control (HC-) and MM-MSCs. NAD +/NADH ratio was decreased in MM-MSCs (n=8) as compared with HC-MSCs (n=4, p In the cell-to-cell contact the gap junction-forming protein CX43 has been found critical for mitochondria uptake in lung and brain injury and it also can regulate CXCL12 secretion by MSCs. We found that MM-MSCs showed a significantly up-regulated CXCL12 expression as compared to HC-MSCs (p In conclusion, we have shown that MM-MSCs are relatively low dependent on mitochondria metabolism and are inclined to transfer mitochondria to MM tumor cells. Furthermore, tumor PCs increase the expression of CX43 in MSCs leading to an increased levels of CXCL12 and stimulation of its corresponding receptor expressed on MM cells. The resulting CX43/CXCL12/CXCR4 interplay enhances mitochondrial trafficking from MSCs to myeloma PCs and can protect cancer cells against anti-myeloma agents. Understanding pro-tumorigenic phenotype of MSCs and mechanisms of adhesion and heterocellular communication favoring their interaction with cancer PCs, will allow to manipulate critical pathways, including CXCL12/CXCR4 axis, thus improving disease outcome. Disclosures Di Raimondo: Pfizer: Honoraria; AbbVie: Honoraria; Bristol Myers Squibb: Honoraria; Jazz Pharmaceutical: Honoraria; Janssen Pharmaceuticals: Honoraria; Amgen: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
20. The Heme Oxygenase-1/Carbon Monoxide Pathway Activates TLR4 Signaling Promoting Bortezomib Resistance in Multiple Myeloma Cells
- Author
-
Grazia Scandura, Daniela Cambria, Enrico La Spina, Concetta Conticello, Giovanni Li Volti, Alessandra Romano, Michelino Di Rosa, Francesco Di Raimondo, Giuseppe Musumeci, Alessandro Barbato, Fabrizio Puglisi, Daniele Tibullo, Giuseppe A. Palumbo, and Cesarina Giallongo
- Subjects
Bortezomib ,Immunology ,Tlr4 signaling ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Heme oxygenase ,chemistry.chemical_compound ,chemistry ,medicine ,Cancer research ,Multiple myeloma ,Carbon monoxide ,medicine.drug - Abstract
Heme oxygenase (HO)-1 catalyzes the conversion of heme to biliverdin, iron an d carbon monoxide. In myeloma plasma cells, its expression has been demonstrated to increase during bortezomib (BTZ) treatment and localize into the nucleus conferring drug resistance. Recently, our group demonstrated that BTZ also induces up-regulation of Toll like receptor 4 (TLR4) which acts as a stress-responsive mechanism protecting mitochondria during BTZ exposure and sustaining mitochondrial metabolism. Since two studies independently demonstrated that both HO-1 and TLR4 protect myeloma cells from BTZ-induced apoptotic signals, specific functional connections between these two proteins were considered herein. MM cell lines were treated with BTZ alone or in combination with TAK-242, a selective inhibitor of TLR4. We observed a significant increase of apoptosis in TAK-242/BTZ treated cells compared to BTZ alone. Drug combination also led to higher mitochondrial depolarization and decreased mitochondrial mass evaluated using flow cytometry. Accordingly, TAK-242/BTZ treatment act ivated mitophagy as demonstrated by evaluating co-localization of the autophagosome marker LC3 with mitochondria using confocal microscopy. Since it is known BTZ treated cells increased HO-1 expression as protective mechanism, we next evaluated if BTZ combination with TAK-242 could affect HO-1 expression. Western blot showed a down-regulation of HO-1 after TAK-242/BTZ treatment. Immunofluorescence analysis confirmed that drug combination decreased nuclear HO-1 and increased its cytoplasmic localization compared to BTZ alone. To address this controversy, we administered tin protoporphyrin (SnPP), a well-characterized HO-1 enzymatic inhibitor, alone or in combination with BTZ. Interestingly, SnPP/BTZ treated cells showed lower expression of TLR4 compared to BTZ treated ones. To better investigate if HO-1 enzymatic activity could regulate TLR4 expression, MM cells were exposed to hemin, an inducer of HO-1. We observed a significant up-regulation of TLR4 and NF-kB nuclear localization. Treating cells with rapid or slowly carbon monoxide-releasing molecules (CORM-3 and CORM-A1), an increase of TLR4 expression was observed after 3h with the consequent activation of p-p38, p-ERK and NF-kB nuclear translocation. Moreover, silencing HO-1 confirmed its role in the regulation of TLR4 expression. Moreover, compared to U266 cells, shHO-1/U266 cells howed higher apoptosis after treatment with BTZ, confirming that HO-1/TLR4 signaling protect MM cells from BTZ-induced apoptosis. Our data demonstrate that a functional regulatory link exists between HO1 and TLR4 which in turn impact on drug response. Specifically, inhibition of HO-1/TLR4 axis augmented cytotoxicity of BTZ against MM cells. In conclusion the HO-1/TLR4 axis is involved in BTZ mediated chemoresistance thus providing an important tool to improve the clinical outcome of MM patients resistant to BTZ. Disclosures Romano: Takeda: Honoraria; Novartis: Honoraria. Puglisi:Amgen: Honoraria. Conticello:Amgen, Takeda, Janssen: Honoraria. Palumbo:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Di Raimondo:Amgen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; GSK: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Amgen, Takeda, Novartis: Honoraria; GILEAD, Incyte: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
21. An Integrative Unbiased Global Analysis of the Metabolic Adaptive Response to Tryptophan Deprivation in Classical Hodgkin Lymphoma
- Author
-
Grazia Scandura, Daniele Tibullo, Cesarina Giallongo, Alessandro Barbato, Alessandra Romano, Enrico La Spina, Francesco Di Raimondo, and Fabrizio Puglisi
- Subjects
Methionine ,Immunology ,Tryptophan ,Cell Biology ,Hematology ,Mitochondrion ,Pentose phosphate pathway ,Biochemistry ,Warburg effect ,chemistry.chemical_compound ,Metabolic pathway ,Tryptophan Metabolite ,chemistry ,NAD+ kinase - Abstract
Background Tryptophan (trp) is an essential aminoacid, required for de novo NAD+ synthesis. Our previous work showed that the microenvironment in classical Hodgkin Lymphoma is characterized by dysfunctional neutrophils and myeloid derived suppressor cells that produce a trp-degrading enzyme indoelamine deoxygenase (IDO-1), lowering the amount of trp. Aims To detect metabolomics changes in human cHL cell lines exposed in vitro to tryptophan deprivation, an amino acid involved in immune dysregulation and generation of anergic and tolerogenic T- cells. Methods In order to better understand the impact of extra-cellular IDO1 increase on the metabolome of human cHL cells, three human cHL cell lines (L428, L540 and KM-H2) were individually cultured with customized complete media or media lacking tryptophan (W0), +10% dyalised fetal bovum serum, in six independent experiments. After 48 hours of culture the cells were collected for global metabolomic analysis, by gas chromatography-mass spectrometry (GC/MS) and liquid chromatography-tandem mass spectrometry (LC/MS/MS) platforms by Metabolon Inc. Following normalization to DNA concentration, log transformation and imputation of missing values, if any, with the minimum observed value for each compound, Welch's two-sample t-test was used to identify biochemicals that differed significantly between experimental groups (Table 1). Results The lack of tryptophan in media had a profound effect on the cell metabolome in 2 cell lines, KMH2 and L428 cells, while L540 cell line was pretty resilient (Table 1). In all cell lines, the removal of tryptophan from the media resulted in significantly lower levels of tryptophan. Kynurenine, the metabolic product of IDO-1 action on tryptophan, was lower in all cells, but did not reach significance in the L540 cells, whereas it trended lower in L428 and was significantly lower in the KMH2 compared to controls. Indolelactate, another major tryptophan metabolite was also significantly lower in the L428 and KMH2 cells lines compared to controls. Glucose uptake and aerobic glycolysis are frequently upregulated in tumor cells to support energy needs and provide biosynthetic precursors (e.g. pentose phosphate pathway intermediates for nucleotide synthesis). Known as the Warburg effect, this process of reliance on glucose for energy results in high levels of lactate production. We found that trp deficiency lead to lower levels of the hexose diphosphates (fructose 1,6-diphosphate/glucose 1,6-diphosphate/myo-inositol diphosphates isobar) and dihydroxyacetone phosphate, suggesting the revert of Warburg effect due to reduced bio-energetic requirements for proliferation. In line with this observation, culture in trp deficient media resulted in increased levels of long chain saturated fatty acids and long chain polyunsaturated fatty acids (PUFAs), suggesting that specific-amino acid deficiency leads to an increase in uptake of free fatty acids from the media, to preserve membrane dynamics. Since prolonged trp deprivation (up to 10 days) delayed cell cycle length without affecting proliferation or changes in intracellular amount of NAD+, we investigated changes in mitochondrial membranes network to explain these findings. Trp deprivation induced the rearrangement of the mitochondrial network at 48 hours, with more fission than fusion, as suggested by increased expression of Fis1 and Drp1 and reduced expression of Tfam and Opa1, without affecting significantly mitochondrial mass and depolarization. This adaptive response was associated to increased oxidative stress, as suggested by of reduced glutathione (GSH) and oxidized glutathione (GSSG) in the L540 and KMH2 cells, depletion of gamma-glutamylcysteine, increased cystine, the oxidative product of cysteine, and methionine sulfoxide (an oxidation product of methionine). Conclusions The removal of trp from L428 and KMH2 resulted in changes in the specific-amino acid related metabolites. The adaptive response to trp-depleted microenvironment can revert the Warburg effect, promoting a shift in the glucose use in the futile attempt to preserve mitochondrial function, and increase oxidative stress. Quantity and function of mitochondria network can play a major role in selecting the fittest clones, a metabolic pathway that should be explored as novel non -synthetic lethal targets. Disclosures Puglisi: Amgen:Honoraria.Di Raimondo:Takeda:Consultancy, Honoraria;GILEAD, Incyte:Research Funding;Amgen, Takeda, Novartis:Honoraria;Celgene:Consultancy, Honoraria;Janssen:Consultancy, Honoraria;GSK:Consultancy, Honoraria;Amgen:Consultancy, Honoraria.Romano:Takeda:Honoraria;Novartis:Honoraria.
- Published
- 2020
- Full Text
- View/download PDF
22. Focus on Osteosclerotic Progression in Primary Myelofibrosis
- Author
-
Grazia Scandura, Michelino Di Rosa, Giuseppe A. Palumbo, Giovanni Li Volti, Alessandra Romano, Enrico La Spina, Vincenzo Bramanti, Mariarita Spampinato, Ilaria Dulcamare, Cesarina Giallongo, Rosalba Parenti, Lucia Longhitano, Nunzio Vicario, Daniele Tibullo, and Roberto Avola
- Subjects
0301 basic medicine ,Chemokine ,bone marrow ,myeloproliferative neoplasm ,lcsh:QR1-502 ,Review ,Bone tissue ,bone ,Biochemistry ,Monocytes ,lcsh:Microbiology ,03 medical and health sciences ,Osteosclerosis ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,Humans ,Myelofibrosis ,Molecular Biology ,Myeloproliferative neoplasm ,biology ,business.industry ,fibrosis ,medicine.disease ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Disease Progression ,primary myelofibrosis ,Cancer research ,biology.protein ,Bone marrow ,business ,Signal Transduction - Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.