Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Genet ; 60(4): 317-326, 2023 04.
Article in English | MEDLINE | ID: mdl-36849229

ABSTRACT

BACKGROUND: Birt-Hogg-Dubé (BHD) syndrome is a rare genetic syndrome caused by pathogenic or likely pathogenic germline variants in the FLCN gene. Patients with BHD syndrome have an increased risk of fibrofolliculomas, pulmonary cysts, pneumothorax and renal cell carcinoma. There is debate regarding whether colonic polyps should be added to the criteria. Previous risk estimates have mostly been based on small clinical case series. METHODS: A comprehensive review was conducted to identify studies that had recruited families carrying pathogenic or likely pathogenic variants in FLCN. Pedigree data were requested from these studies and pooled. Segregation analysis was used to estimate the cumulative risk of each manifestation for carriers of FLCN pathogenic variants. RESULTS: Our final dataset contained 204 families that were informative for at least one manifestation of BHD (67 families informative for skin manifestations, 63 for lung, 88 for renal carcinoma and 29 for polyps). By age 70 years, male carriers of the FLCN variant have an estimated 19% (95% CI 12% to 31%) risk of renal tumours, 87% (95% CI 80% to 92%) of lung involvement and 87% (95% CI 78% to 93%) of skin lesions, while female carriers had an estimated 21% (95% CI 13% to 32%) risk of renal tumours, 82% (95% CI 73% to 88%) of lung involvement and 78% (95% CI 67% to 85%) of skin lesions. The cumulative risk of colonic polyps by age 70 years old was 21% (95% CI 8% to 45%) for male carriers and 32% (95% CI 16% to 53%) for female carriers. CONCLUSIONS: These updated penetrance estimates, based on a large number of families, are important for the genetic counselling and clinical management of BHD syndrome.


Subject(s)
Birt-Hogg-Dube Syndrome , Carcinoma, Renal Cell , Colonic Polyps , Kidney Neoplasms , Humans , Male , Female , Aged , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/pathology , Penetrance , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Kidney Neoplasms/epidemiology , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/genetics
2.
Semin Cell Dev Biol ; 98: 90-97, 2020 02.
Article in English | MEDLINE | ID: mdl-31091466

ABSTRACT

The efficacy of metformin in treating cancer has been extensively investigated since epidemiologic studies associated this anti-diabetic drug with a lower risk of cancer incidence. Since tumors are complex systems, in which cancer cells coexist and interact with several different types of non-malignant cells, it is not surprising that anti-cancer drugs affect not only cancer cells, but also the abundance and functions of cells of the tumor microenvironment. Recent years have seen a wide collection of reports showing how metformin, as well as other complex I inhibitors, may influence cancer progression by modulating the phenotype of non-transformed cells in a tumor. In this review, we particularly focus on the effect of metformin on angiogenesis, cancer-associated fibroblasts, tumor-associated macrophages and cancer immunosuppression.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Macrophages/drug effects , Macrophages/pathology , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
3.
Semin Cell Dev Biol ; 98: 26-33, 2020 02.
Article in English | MEDLINE | ID: mdl-31175937

ABSTRACT

The thriving field that constitutes cancer metabolism has unveiled some groundbreaking facts over the past two decades, at the heart of which is the TCA cycle and its intermediates. As such and besides its metabolic role, α-ketoglutarate was shown to withstand a wide range of physiological reactions from protection against oxidative stress, collagen and bone maintenance to development and immunity. Most importantly, it constitutes the rate-limiting substrate of 2-oxoglutarate-dependent dioxygenases family enzymes, which are involved in hypoxia sensing and in the shaping of cellular epigenetic landscape, two major drivers of oncogenic transformation. Based on literature reports, we hereby review the benefits of this metabolite as a possible novel adjuvant therapeutic opportunity to target tumor progression. This article is part of the special issue "Mitochondrial metabolic alterations in cancer cells and related therapeutic targets".


Subject(s)
Ketoglutaric Acids/metabolism , Neoplasms/metabolism , Animals , Disease Progression , Humans , Mitochondria/metabolism , Neoplasms/pathology
4.
Nucleic Acids Res ; 47(D1): D1202-D1210, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30371888

ABSTRACT

Interest in human mitochondrial genetic data is constantly increasing among both clinicians and researchers, due to the involvement of mitochondrial DNA (mtDNA) in a number of physiological and pathological processes. Thanks to new sequencing technologies and modern databases, the large amount of information on mtDNA variability may be exploited to gain insights into the relationship between mtDNA variants, phenotypes and diseases. To facilitate this process, we have developed the HmtVar resource, a variant-focused database that allows the exploration of a dataset of over 40 000 human mitochondrial variants. Mitochondrial variation data, initially gathered from the HmtDB platform, are integrated with in-house pathogenicity assessments based on various evaluation criteria and with a set of additional annotations from third-party resources. The result is a comprehensive collection of information of crucial importance for human mitochondrial variation studies and investigation of common and rare diseases in which the mitochondrion may be involved. HmtVar is accessible at https://www.hmtvar.uniba.it and data may be retrieved using either a web interface through the Query page or a state-of-the-art API for programmatic access.


Subject(s)
Computational Biology/methods , DNA, Mitochondrial/genetics , Databases, Genetic , Genetic Variation , Genome, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Databases, Nucleic Acid , Genes, Mitochondrial/genetics , Genetic Predisposition to Disease/genetics , Genomics/methods , Humans , Internet , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Phenotype
5.
Int J Mol Sci ; 22(11)2021 May 29.
Article in English | MEDLINE | ID: mdl-34072463

ABSTRACT

The NBN gene has been included in breast cancer (BC) multigene panels based on early studies suggesting an increased BC risk for carriers, though not confirmed by recent research. To evaluate the impact of NBN analysis, we assessed the results of NBN sequencing in 116 BRCA-negative BC patients and reviewed the literature. Three patients (2.6%) carried potentially relevant variants: two, apparently unrelated, carried the frameshift variant c.156_157delTT and another one the c.628G>T variant. The latter was subsequently found in 4/1390 (0.3%) BC cases and 8/1580 (0.5%) controls in an independent sample, which, together with in silico predictions, provided evidence against its pathogenicity. Conversely, the rare c.156_157delTT variant was absent in the case-control set; moreover, a 50% reduction of NBN expression was demonstrated in one carrier. However, in one family it failed to co-segregate with BC, while the other carrier was found to harbor also a probably pathogenic TP53 variant that may explain her phenotype. Therefore, the c.156_157delTT, although functionally deleterious, was not supported as a cancer-predisposing defect. Pathogenic/likely pathogenic NBN variants were detected by multigene panels in 31/12314 (0.25%) patients included in 15 studies. The risk of misinterpretation of such findings is substantial and supports the exclusion of NBN from multigene panels.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Nuclear Proteins/genetics , Adult , Alleles , Case-Control Studies , DNA Mutational Analysis , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies/methods , Genetic Testing , Genotype , Haplotypes , Humans , Pedigree
6.
Proc Natl Acad Sci U S A ; 114(43): E9066-E9075, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073103

ABSTRACT

The horizontal transfer of mtDNA and its role in mediating resistance to therapy and an exit from dormancy have never been investigated. Here we identified the full mitochondrial genome in circulating extracellular vesicles (EVs) from patients with hormonal therapy-resistant (HTR) metastatic breast cancer. We generated xenograft models of HTR metastatic disease characterized by EVs in the peripheral circulation containing mtDNA. Moreover, these human HTR cells had acquired host-derived (murine) mtDNA promoting estrogen receptor-independent oxidative phosphorylation (OXPHOS). Functional studies identified cancer-associated fibroblast (CAF)-derived EVs (from patients and xenograft models) laden with whole genomic mtDNA as a mediator of this phenotype. Specifically, the treatment of hormone therapy (HT)-naive cells or HT-treated metabolically dormant populations with CAF-derived mtDNAhi EVs promoted an escape from metabolic quiescence and HTR disease both in vitro and in vivo. Moreover, this phenotype was associated with the acquisition of EV mtDNA, especially in cancer stem-like cells, expression of EV mtRNA, and restoration of OXPHOS. In summary, we have demonstrated that the horizontal transfer of mtDNA from EVs acts as an oncogenic signal promoting an exit from dormancy of therapy-induced cancer stem-like cells and leading to endocrine therapy resistance in OXPHOS-dependent breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , DNA, Mitochondrial/metabolism , Drug Resistance, Neoplasm/genetics , Exosomes/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , DNA, Mitochondrial/genetics , Female , Fibroblasts/pathology , Gene Transfer, Horizontal , Genome, Mitochondrial/genetics , Humans , MCF-7 Cells , NADH Dehydrogenase/genetics , Oxidative Phosphorylation , Receptors, Estrogen/metabolism , Xenograft Model Antitumor Assays
7.
Neurobiol Dis ; 124: 14-28, 2019 04.
Article in English | MEDLINE | ID: mdl-30389403

ABSTRACT

Spinocerebellar ataxia 28 is an autosomal dominant neurodegenerative disorder caused by missense mutations affecting the proteolytic domain of AFG3L2, a major component of the mitochondrial m-AAA protease. However, little is known of the underlying pathogenetic mechanisms or how to treat patients with SCA28. Currently available Afg3l2 mutant mice harbour deletions that lead to severe, early-onset neurological phenotypes that do not faithfully reproduce the late-onset and slowly progressing SCA28 phenotype. Here we describe production and detailed analysis of a new knock-in murine model harbouring an Afg3l2 allele carrying the p.Met665Arg patient-derived mutation. Heterozygous mutant mice developed normally but adult mice showed signs of cerebellar ataxia detectable by beam test. Although cerebellar pathology was negative, electrophysiological analysis showed a trend towards increased spontaneous firing in Purkinje cells from heterozygous mutants with respect to wild-type controls. As homozygous mutants died perinatally with evidence of cardiac atrophy, for each genotype we generated mouse embryonic fibroblasts (MEFs) to investigate mitochondrial function. MEFs from mutant mice showed altered mitochondrial bioenergetics, with decreased basal oxygen consumption rate, ATP synthesis and mitochondrial membrane potential. Mitochondrial network formation and morphology was altered, with greatly reduced expression of fusogenic Opa1 isoforms. Mitochondrial alterations were also detected in cerebella of 18-month-old heterozygous mutants and may be a hallmark of disease. Pharmacological inhibition of de novo mitochondrial protein translation with chloramphenicol caused reversal of mitochondrial morphology in homozygous mutant MEFs, supporting the relevance of mitochondrial proteotoxicity for SCA28 pathogenesis and therapy development.


Subject(s)
ATP-Dependent Proteases/genetics , ATPases Associated with Diverse Cellular Activities/genetics , Disease Models, Animal , Mitochondria/metabolism , Spinocerebellar Ataxias/congenital , Animals , Female , Gene Knock-In Techniques , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mutation, Missense , Purkinje Cells/physiology , Purkinje Cells/ultrastructure , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
8.
Hum Mol Genet ; 26(15): 2961-2974, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28486623

ABSTRACT

Development of chemoresistance is a cogent clinical issue in oncology, whereby combination of anticancer drugs is usually preferred also to enhance efficacy. Paclitaxel (PTX), combined with carboplatin, represents the standard first-line chemotherapy for different types of cancers. We here depict a double-edge role of mitochondrial DNA (mtDNA) mutations induced in cancer cells after treatment with platinum. MtDNA mutations were positively selected by PTX, and they determined a decrease in the mitochondrial respiratory function, as well as in proliferative and tumorigenic potential, in terms of migratory and invasive capacity. Moreover, cells bearing mtDNA mutations lacked filamentous tubulin, the main target of PTX, and failed to reorient the Golgi body upon appropriate stimuli. We also show that the bioenergetic and cytoskeletal phenotype were transferred along with mtDNA mutations in transmitochondrial hybrids, and that this also conferred PTX resistance to recipient cells. Overall, our data show that platinum-induced deleterious mtDNA mutations confer resistance to PTX, and confirm what we previously reported in an ovarian cancer patient treated with carboplatin and PTX who developed a quiescent yet resistant tumor mass harboring mtDNA mutations.


Subject(s)
DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Paclitaxel/metabolism , Antineoplastic Agents/pharmacology , Carboplatin/metabolism , Cell Line, Tumor , Cytoskeleton/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Mutation/drug effects , Ovarian Neoplasms/genetics , Platinum , Tubulin/drug effects , Tubulin/genetics , Tubulin/metabolism
9.
Ann Surg ; 270(6): 969-975, 2019 12.
Article in English | MEDLINE | ID: mdl-30672801

ABSTRACT

OBJECTIVE: To evaluate the effectiveness of a program to reduce work-related musculoskeletal disorders (WRMSD) among surgeons. BACKGROUND: Surgeons are at high risk of WRMSD due to many physical and psychosocial factors. METHODS: This study is a multicenter randomized clinical trial (UMIN000028557) conducted from January to August 2015. Following cluster randomization by surgical division, surgeons were allocated to 2 groups. The NPP group (No Preventive Program) underwent no intervention, while the PP group (Preventive Program) followed ergonomic principles in the operating room and specific physical exercises supervised by a physical therapist. A multiple logistic regression was performed to identify baseline WRMSD risk factors. WRMSD assessment was based on 1 ad hoc and 3 validated questionnaires: Nordic Musculoskeletal Questionnaire (NMQ), Numerical Rating Scale (NRS), and Short Form 36 Health Survey (SF-36). Follow-up was planned after 3 and 6 months. RESULTS: One hundred forty-one surgeons matched the inclusion criteria and were randomized in the PP (n = 65) and NPP (n = 76) groups. At the initial analysis, physical activity was the only modifiable independent risk factor for WRMSD (OR, 2.44; P = 0.05). The PP group showed a significant improvement in the item "General Health" (GH) regarding quality of life at 3 (NPP: 70.5 ±â€Š15.2 vs PP: 75.9 ±â€Š14.1; P = 0.04) and 6 months (70.6 ±â€Š13.4 vs 75.3 ±â€Š13.0; P = 0.04). The PP group had a significant reduction of low back pain (66.2% vs 50.0%; P = 0.04) and analgesic consumption (30.9% vs 15.5%; P = 0.03) after 6 months. CONCLUSIONS: This study demonstrated the effectiveness of a global program based on the application of ergonomics in the operating room and specific physical exercises.


Subject(s)
Ergonomics , Musculoskeletal Diseases/prevention & control , Occupational Diseases/prevention & control , Preventive Health Services , Surgeons , Adult , Female , Humans , Male , Middle Aged , Musculoskeletal Diseases/diagnosis , Musculoskeletal Diseases/etiology , Occupational Diseases/diagnosis , Occupational Diseases/etiology , Risk Factors
10.
Nucleic Acids Res ; 45(D1): D698-D706, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27899581

ABSTRACT

The HmtDB resource hosts a database of human mitochondrial genome sequences from individuals with healthy and disease phenotypes. The database is intended to support both population geneticists as well as clinicians undertaking the task to assess the pathogenicity of specific mtDNA mutations. The wide application of next-generation sequencing (NGS) has provided an enormous volume of high-resolution data at a low price, increasing the availability of human mitochondrial sequencing data, which called for a cogent and significant expansion of HmtDB data content that has more than tripled in the current release. We here describe additional novel features, including: (i) a complete, user-friendly restyling of the web interface, (ii) links to the command-line stand-alone and web versions of the MToolBox package, an up-to-date tool to reconstruct and analyze human mitochondrial DNA from NGS data and (iii) the implementation of the Reconstructed Sapiens Reference Sequence (RSRS) as mitochondrial reference sequence. The overall update renders HmtDB an even more handy and useful resource as it enables a more rapid data access, processing and analysis. HmtDB is accessible at http://www.hmtdb.uniba.it/.


Subject(s)
DNA, Mitochondrial , Databases, Nucleic Acid , Genome, Mitochondrial , Genomics/methods , Haplotypes , Mitochondria/genetics , Humans , Search Engine , Software , Web Browser
11.
Hum Mutat ; 39(1): 92-102, 2018 01.
Article in English | MEDLINE | ID: mdl-28967163

ABSTRACT

Respiratory complex III (CIII) is the first enzymatic bottleneck of the mitochondrial respiratory chain both in its native dimeric form and in supercomplexes. The mammalian CIII comprises 11 subunits among which cytochrome b is central in the catalytic core, where oxidation of ubiquinol occurs at the Qo site. The Qo- or PEWY-motif of cytochrome b is the most conserved through species. Importantly, the highly conserved glutamate at position 271 (Glu271) has never been studied in higher eukaryotes so far and its role in the Q-cycle remains debated. Here, we showed that the homoplasmic m.15557G > A/MT-CYB, which causes the p.Glu271Lys amino acid substitution predicted to dramatically affect CIII, induces a mild mitochondrial dysfunction in human transmitochondrial cybrids. Indeed, we found that the severity of such mutation is mitigated by the proper assembly of CIII into supercomplexes, which may favor an optimal substrate channeling and buffer superoxide production in vitro.


Subject(s)
Alleles , Cytochromes b/genetics , Genetic Association Studies , Mutation , Phenotype , Adenosine Triphosphate , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Cell Survival/genetics , Conserved Sequence , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Energy Metabolism , Humans , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism
12.
BMC Cancer ; 18(1): 7, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29295713

ABSTRACT

BACKGROUND: Extra-abdominal metastases in low grade endometrial carcinoma are rare events. Inguinal lymphatic spread occurs usually in advanced disease and is associated with abdominal lymph nodes involvement. To our knowledge, isolated inguinal lymph node metastases in patients with early endometrial carcinoma have never been described thus far. CASE PRESENTATION: We present an uncommon case of inguinal lymph node metastasis in a 51-year old patient with early endometrial disease without other metastatic involvement. The metastatic loci were analyzed with the recently validated method of mitochondrial DNA sequencing to demonstrate clonality of the lesions. CONCLUSIONS: We describe the first case of inguinal metastasis from intramucous endometrial carcinoma; this case confirms the unpredictable spread of endometrial neoplasia and the importance of both patient's history and physical examination in good clinical practice.


Subject(s)
Adenocarcinoma/secondary , Endometrial Neoplasms/pathology , Inguinal Canal/pathology , Lymph Nodes/pathology , Adenocarcinoma/genetics , Adenocarcinoma/surgery , DNA, Mitochondrial/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/surgery , Female , Humans , Inguinal Canal/surgery , Lymph Nodes/surgery , Middle Aged , Neoplasm Grading
13.
Int J Mol Sci ; 19(7)2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30011887

ABSTRACT

In the event of multiple synchronous gynecological lesions, a fundamental piece of information to determine patient management, prognosis, and therapeutic regimen choice is whether the simultaneous malignancies arise independently or as a result of metastatic dissemination. An example of synchronous primary tumors of the female genital tract most frequently described are ovarian and endometrial cancers. Surgical findings and histopathological examination aimed at resolving this conundrum may be aided by molecular analyses, although they are too often inconclusive. High mitochondrial DNA (mtDNA) variability and its propensity to accumulate mutations has been proposed by our group as a tool to define clonality. We showed mtDNA sequencing to be informative in synchronous primary ovarian and endometrial cancer, detecting tumor-specific mutations in both lesions, ruling out independence of the two neoplasms, and indicating clonality. Furthermore, we tested this method in another frequent simultaneously detected gynecological lesion type, borderline ovarian cancer and their peritoneal implants, which may be monoclonal extra-ovarian metastases or polyclonal independent masses. The purpose of this review is to provide an update on the potential use of mtDNA sequencing in distinguishing independent and metastatic lesions in gynecological cancers, and to compare the efficiency of molecular analyses currently in use with this novel method.


Subject(s)
DNA, Mitochondrial/genetics , Genital Neoplasms, Female/diagnosis , Genital Neoplasms, Female/genetics , Sequence Analysis, DNA/methods , Diagnosis, Differential , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Female , Genital Neoplasms, Female/classification , Humans , Mutation , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Sensitivity and Specificity
14.
Int J Mol Sci ; 19(3)2018 Mar 07.
Article in English | MEDLINE | ID: mdl-29518970

ABSTRACT

Mammalian respiratory complex I (CI) biogenesis requires both nuclear and mitochondria-encoded proteins and is mostly organized in respiratory supercomplexes. Among the CI proteins encoded by the mitochondrial DNA, NADH-ubiquinone oxidoreductase chain 1 (ND1) is a core subunit, evolutionary conserved from bacteria to mammals. Recently, ND1 has been recognized as a pivotal subunit in maintaining the structural and functional interaction among the hydrophilic and hydrophobic CI arms. A critical role of human ND1 both in CI biogenesis and in the dynamic organization of supercomplexes has been depicted, although the proof of concept is still missing and the critical amount of ND1 protein necessary for a proper assembly of both CI and supercomplexes is not defined. By exploiting a unique model in which human ND1 is allotopically re-expressed in cells lacking the endogenous protein, we demonstrated that the lack of this protein induces a stall in the multi-step process of CI biogenesis, as well as the alteration of supramolecular organization of respiratory complexes. We also defined a mutation threshold for the m.3571insC truncative mutation in mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (MT-ND1), below which CI and its supramolecular organization is recovered, strengthening the notion that a certain amount of human ND1 is required for CI and supercomplexes biogenesis.


Subject(s)
Alleles , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Mutation , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Cell Respiration , DNA, Mitochondrial/genetics , Electron Transport Complex I/metabolism , Mitochondria/genetics , Mitochondria/metabolism , NADH Dehydrogenase/metabolism , Oxygen Consumption , Protein Binding , Structure-Activity Relationship
15.
Biochim Biophys Acta Bioenerg ; 1858(8): 582-590, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28213331

ABSTRACT

Energy homeostasis is pivotal for cell fate since metabolic regulation, cell proliferation and death are strongly dependent on the balance between catabolic and anabolic pathways. In particular, metabolic and energetic changes have been observed in cancer cells even before the discovery of oncogenes and tumor suppressors, but have been neglected for a long time. Instead, during the past 20years a renaissance of the study of tumor metabolism has led to a revised and more accurate sight of the metabolic landscape of cancer cells. In this scenario, genetic, biochemical and clinical evidences place mitochondria as key actors in cancer metabolic restructuring, not only because there are energy and biosynthetic intermediates manufacturers, but also because occurrence of mutations in metabolic enzymes encoded by both nuclear and mitochondrial DNA has been associated to different types of cancer. Here we provide an overview of the possible mechanisms modulating mitochondrial energy production and homeostasis in the intriguing scenario of neoplastic cells, focusing on the double-edged role of 5'-AMP activated protein kinase in cancer metabolism. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Energy Metabolism , Mitochondria/metabolism , Neoplasms/metabolism , AMP-Activated Protein Kinases/physiology , Adenosine Triphosphate/metabolism , Animals , Disease Progression , Genes, Mitochondrial , Homeostasis , Humans , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Models, Biological , Neoplasm Proteins/physiology , Neoplasms/pathology , Neoplasms, Experimental/metabolism , Tumor Suppressor Proteins/physiology
16.
Biochim Biophys Acta Bioenerg ; 1858(8): 591-601, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28115060

ABSTRACT

Oncocytic tumors are a peculiar subset of human neoplasms in which mitochondria have been proven to have a prominent role. A number of paradoxes render these clinical entities interesting from the translational research point of view. Most oncocytic tumors are generally metabolically constrained due to the impaired respiratory capacity and lack of the ability to respond to hypoxia, yet they maintain features that allow them to strive and persist in an indolent form. Their unique molecular and metabolic characteristics are an object of investigation that may reveal novel ways for therapeutic strategies based on metabolic targeting. With this aim in mind, we here examine the current knowledge on oncocytomas and delve into the molecular causes and consequences that revolve around the oncocytic phenotype, to understand whether we can learn to design therapies from the dissection of benign neoplasms. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Adenoma, Oxyphilic/metabolism , Mitochondria/metabolism , Adenoma, Oxyphilic/drug therapy , Adenoma, Oxyphilic/genetics , DNA, Mitochondrial/genetics , Disease Progression , Electron Transport Complex I/metabolism , Energy Metabolism , Genes, Neoplasm , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Models, Biological , Molecular Targeted Therapy , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/metabolism , Organelle Biogenesis , Phenotype
17.
Mol Cancer ; 16(1): 47, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28241835

ABSTRACT

Borderline ovarian tumors are rare low malignant potential neoplasms characterized by the absence of stromal invasion, whose main prognostic factors are stage and type of peritoneal implants. The latter are defined as invasive when cell proliferation invades the underlying tissue (peritoneal surface, omentum and intestinal wall), or noninvasive. It is still unknown if these implants are metastatic spread from the primary ovarian mass or a neoplastic transformation de novo of the peritoneal surface. Mitochondrial DNA sequencing was performed to assess clonality in eight patients presenting both borderline ovarian tumors and implants. In 37.5% of the cases, the same mitochondrial DNA mutation was present in both borderline ovarian tumors and the peritoneal implant, being this evidence that implants may arise as a consequence of a spread from a single ovarian site.


Subject(s)
Clonal Evolution , DNA, Mitochondrial , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/surgery , Prognosis , Sequence Analysis, DNA
18.
Mol Cancer ; 16(1): 67, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28327152

ABSTRACT

BACKGROUND: TRIM8 plays a key role in controlling the p53 molecular switch that sustains the transcriptional activation of cell cycle arrest genes and response to chemotherapeutic drugs. The mechanisms that regulate TRIM8, especially in cancers like clear cell Renal Cell Carcinoma (ccRCC) and colorectal cancer (CRC) where it is low expressed, are still unknown. However, recent studies suggest the potential involvement of some microRNAs belonging to miR-17-92 and its paralogous clusters, which could include TRIM8 in a more complex pathway. METHODS: We used RCC and CRC cell models for in-vitro experiments, and ccRCC patients and xenograft transplanted mice for in vivo assessments. To measure microRNAs levels we performed RT-qPCR, while steady-states of TRIM8, p53, p21 and N-MYC were quantified at protein level by Western Blotting as well as at transcript level by RT-qPCR. Luciferase reporter assays were performed to assess the interaction between TRIM8 and specific miRNAs, and the potential effects of this interaction on TRIM8 expression. Moreover, we treated our cell models with conventional chemotherapeutic drugs or tyrosine kinase inhibitors, and measured their response in terms of cell proliferation by MTT and colony suppression assays. RESULTS: We showed that TRIM8 is a target of miR-17-5p and miR-106b-5p, whose expression is promoted by N-MYC, and that alterations of their levels affect cell proliferation, acting on the TRIM8 transcripts stability, as confirmed in ccRCC patients and cell lines. In addition, reducing the levels of miR-17-5p/miR-106b-5p, we increased the chemo-sensitivity of RCC/CRC-derived cells to anti-tumour drugs used in the clinic. Intriguingly, this occurs, on one hand, by recovering the p53 tumour suppressor activity in a TRIM8-dependent fashion and, on the other hand, by promoting the transcription of miR-34a that turns off the oncogenic action of N-MYC. This ultimately leads to cell proliferation reduction or block, observed also in colon cancer xenografts overexpressing TRIM8. CONCLUSIONS: In this paper we provided evidence that TRIM8 and its regulators miR-17-5p and miR-106b-5 participate to a feedback loop controlling cell proliferation through the reciprocal modulation of p53, miR-34a and N-MYC. Our experiments pointed out that this axis is pivotal in defining drug responsiveness of cancers such ccRCC and CRC.


Subject(s)
Carrier Proteins/genetics , Drug Resistance, Neoplasm/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nerve Tissue Proteins/genetics , Tumor Suppressor Protein p53/genetics , 3' Untranslated Regions , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carrier Proteins/metabolism , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , RNA Interference , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
19.
Biochem Biophys Res Commun ; 491(1): 85-90, 2017 09 09.
Article in English | MEDLINE | ID: mdl-28698145

ABSTRACT

Pathogenic mtDNA mutations associated with alterations of respiratory complex I, mitochondrial proliferation (oncocytic-like phenotype) and increase in antioxidant response were previously reported in type I endometrial carcinoma (EC). To evaluate whether in the presence of pathogenic mtDNA mutations other mitochondrial adaptive processes are triggered by cancer cells, the expression level of proteins involved in mitochondrial dynamics, mitophagy, proteolysis and apoptosis were evaluated in type I ECs harboring pathogenic mtDNA mutations and complex I deficiency. An increase in the fission protein Drp1, in the mitophagy protein BNIP3, in the mitochondrial protease CLPP, in the antioxidant and anti-apoptotic protein ALR and in Bcl-2 as well as a decrease in the fusion protein Mfn2 were found in cancer compared to matched non malignant tissue. Moreover, the level of these proteins was measured in type I EC, in hyperplastic (the premalignant form) and in non malignant tissues to verify whether the altered expression of these proteins is a common feature of endometrial cancer and of hyperplastic tissue. This analysis confirmed in type I EC samples, but not in hyperplasia, an alteration of the expression level of these proteins. These results suggest that in this cancer mitochondrial fission, antioxidant and anti-apoptotic response may be activated, as well as the discharge of damaged mitochondrial proteins as adaptation processes to mitochondrial dysfunction.


Subject(s)
Electron Transport Complex I/deficiency , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Mitochondria/metabolism , Mitophagy , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Electron Transport Complex I/genetics , Female , Humans , Mitochondrial Proteins , Neoplasm Proteins , Proteolysis , Tumor Cells, Cultured
20.
Hum Mol Genet ; 23(6): 1453-66, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24163135

ABSTRACT

Mitochondrial DNA mutations are currently investigated as modifying factors impinging on tumor growth and aggressiveness, having been found in virtually all cancer types and most commonly affecting genes encoding mitochondrial complex I (CI) subunits. However, it is still unclear whether they exert a pro- or anti-tumorigenic effect. We here analyzed the impact of three homoplasmic mtDNA mutations (m.3460G>A/MT-ND1, m.3571insC/MT-ND1 and m.3243A>G/MT-TL1) on osteosarcoma progression, chosen since they induce different degrees of oxidative phosphorylation impairment. In fact, the m.3460G>A/MT-ND1 mutation caused only a reduction in CI activity, whereas the m.3571insC/MT-ND1 and the m.3243A>G/MT-TL1 mutations induced a severe structural and functional CI alteration. As a consequence, this severe CI dysfunction determined an energetic defect associated with a compensatory increase in glycolytic metabolism and AMP-activated protein kinase activation. Osteosarcoma cells carrying such marked CI impairment displayed a reduced tumorigenic potential both in vitro and in vivo, when compared with cells with mild CI dysfunction, suggesting that mtDNA mutations may display diverse impact on tumorigenic potential depending on the type and severity of the resulting oxidative phosphorylation dysfunction. The modulation of tumor growth was independent from reactive oxygen species production but correlated with hypoxia-inducible factor 1α stabilization, indicating that structural and functional integrity of CI and oxidative phosphorylation are required for hypoxic adaptation and tumor progression.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Energy Metabolism , NADH Dehydrogenase/metabolism , Osteosarcoma/genetics , RNA, Transfer/genetics , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Disease Progression , Electron Transport Complex I/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutagenesis, Insertional , NADH Dehydrogenase/genetics , Osteosarcoma/pathology , Oxidative Phosphorylation , Point Mutation , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL