1
|
Dwivedi S, Gaur VK, Gupta J. Ecotoxicological impact of succinate dehydrogenase inhibitor (SDHI) fungicides on non-targeted organisms: a review. ECOTOXICOLOGY (LONDON, ENGLAND) 2025; 34:341-361. [PMID: 39843715 DOI: 10.1007/s10646-024-02849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/29/2024] [Indexed: 01/24/2025]
Abstract
As the global population continues to grow, the use of pesticides to increase food production is projected to escalate. Pesticides are critical in plant protection, offering a powerful defense against fungal diseases such as apple scab, leaf spot, sclerotinia rot, damping off, sheath blight, and root rot, which threaten crops like cereals, corn, cotton, soybean, sugarcane, tuberous vegetables, and ornamentals. Succinate Dehydrogenase Inhibitor (SDHI) fungicides represent a novel class essential for controlling fungal pathogens and bolstering food security. However, the impact of SDHIs on non-target organisms, including freshwater and terrestrial invertebrates, crustaceans, and oligochaetes, remains insufficiently understood. Empirical studies indicate that SDHIs can induce mortality, mitochondrial dysfunction, oxidative stress, and developmental delays in non-target organims. Additionally, the environmental persistence of these compounds raises concerns about their potential for ecological disruption. The effects of SDHIs on pollinating species and the possible transgenerational transmission of harmful effects warrant further investigation. Comprehensive transcriptomic analyses are necessary to elucidate the molecular disturbances and adverse outcome pathways triggered by SDHIs. Furthermore, there are emerging concerns about the endocrine-disrupting potential of SDHIs in aquatic organisms. For the first time, this review aims to synthesize existing knowledge on the ecotoxicological impacts of SDHIs on non-target organisms and identify critical research directions to address the ecological challenges posed by their use.
Collapse
Affiliation(s)
- Shreya Dwivedi
- Amity Institute of Environmental Sciences, Amity University, Sector-125, Noida, 201301, Uttar Pradesh, India
- Ecotoxicology Laboratory, Toxicology Department, Institute for Industrial Research & Toxicology, F-209, UPSIDC, Industrial Area, MG Road, Ghaziabad, 201013, Uttar Pradesh, India
| | - Vivek Kumar Gaur
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, 201301, Uttar Pradesh, India
| | - Juhi Gupta
- Amity Institute of Environmental Sciences, Amity University, Sector-125, Noida, 201301, Uttar Pradesh, India.
| |
Collapse
|
2
|
Yong L, Fang Y, Jin L, Zhang X, Luis MA, Lin X, Tang S, Cai F. Unveiling the hidden role of SDHA in breast cancer proliferation: a novel therapeutic avenue. Cancer Cell Int 2025; 25:108. [PMID: 40119440 PMCID: PMC11927305 DOI: 10.1186/s12935-025-03746-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND We observed an increased presence of succinate dehydrogenase complex subunit A (SDHA), a mitochondrial enzyme, in breast cancer (BC), which contributes to its proliferation. While SDHA deficiency has been extensively researched in rare disorders, the upregulation of SDHA and its impact on BC remain understudied. The aim of this study is to investigate the role of SDHA in BC. METHODS The mRNA expression of SDHA was analyzed from TCGA, clinical BC tissues and various BC cell lines via qPCR. Immunohistochemistry was also applied to detect the SDHA expression. Our study investigated the functional outcomes of SDHA overexpression and knockdown in BC utilizing clinical BC tissues from patients and various BC cell lines (MDA-MB-453, MDA-MB-468, SKBR3, and MCF-7). Multiple web platforms and software tools, including R, HPA and TISIDB, were employed to perform comprehensive data analysis. SDHA overexpression and siSDHA were transiently transfected into the cancer cells separately to assess expression levels, cellular proliferation, and migration dynamics through colony formation assay, CCK8 assay, wound-healing analysis. RESULTS We found that the mRNA expression level of SDHA was higher in cancer tissues or cells than in non-cancerous tissues or mammary epithelial cell in TCGA dataset, BC clinical specimens and BC cell lines, respectively. High SDHA expression was associated with poor overall survival (OS, p = 0.016) and disease specific survival (DSS, p = 0.024) in BC patients. Besides, our findings revealed MDA-MB-468, SKBR3 and MCF-7 cells transfected with siSDHA exhibited significantly reduced proliferation and migration capabilities. Conversely, the proliferation and migration abilities of these BC cells significantly increased when transfected with SDHA overexpression. CONCLUSIONS In conclusion, this study highlights the previously underestimated role of SDHA in BC proliferation, presenting a novel avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Liyun Yong
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China
| | - Yuan Fang
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China
| | - Lingli Jin
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China
| | - Xiuqin Zhang
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China
| | - Manuel A Luis
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China
| | - Xiaoyan Lin
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China.
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai, 200065, China.
| |
Collapse
|
3
|
Hu M, Wang XT, Xiao XX, Rao Q, Fan LF, Yue JQ. Renal cell carcinoma with succinate dehydrogenase A mutation: A case report and literature review. Oncol Lett 2025; 29:84. [PMID: 39664613 PMCID: PMC11632410 DOI: 10.3892/ol.2024.14830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is a rare subtype of RCC characterized by the presence of a germline mutation in one of the four subunits of the SDH enzyme complex (SDHA, SDHB, SDHC and SDHD). Together with a somatic second hit, these variants lead to the loss of function of the SDH complex. SDH-deficient RCC associated with SDHA mutation is a rare condition; to the best of our knowledge, there have been only four patients reported in the literature. The present study describes the case of a 22-year-old female patient with RCC associated with SDHA gene mutation. Next-generation sequencing and Sanger sequencing identified a novel heterozygous frameshift variant (NM_004168.4: c.992_999dup) in the SDHA gene. In the literature, this mutation has not previously been reported to be associated with RCC. The present description of a patient with a heterozygous SDHA frameshift variant expands the phenotypic spectrum of the SDHA gene, and provides further clinical, morphological and molecular data of SDHA-deficient RCC.
Collapse
Affiliation(s)
- Min Hu
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Xiao-Tong Wang
- Department of Pathology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xue-Xue Xiao
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Li-Fang Fan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jun-Qiu Yue
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
4
|
Wang P, Chen LL, Xiong Y, Ye D. Metabolite regulation of epigenetics in cancer. Cell Rep 2024; 43:114815. [PMID: 39368084 DOI: 10.1016/j.celrep.2024.114815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/17/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024] Open
Abstract
The catalytic activity of most epigenetic enzymes requires a metabolite produced by central carbon metabolism as a cofactor or (co-)substrate. The concentrations of these metabolites undergo dynamic changes in response to nutrient levels and environmental conditions, reprogramming metabolic processes and epigenetic landscapes. Abnormal accumulations of epigenetic modulatory metabolites resulting from mutations in metabolic enzymes contribute to tumorigenesis. In this review, we first present the concept that metabolite regulation of gene expression represents an evolutionarily conserved mechanism from prokaryotes to eukaryotes. We then review how individual metabolites affect epigenetic enzymes and cancer development. Lastly, we discuss the advancement of and opportunity for therapeutic targeting of metabolite-epigenetic regulation in cancer therapy.
Collapse
Affiliation(s)
- Pu Wang
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei-Lei Chen
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yue Xiong
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Cullgen, Inc., 12671 High Bluff Drive, San Diego, CA 92130, USA.
| | - Dan Ye
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
5
|
潘 秀, 卫 昱, 隋 晓, 尹 晓, 郑 林, 曾 浩, 周 桥, 陈 铌. [Succinate Dehydrogenase-Deficient Renal Cell Carcinoma: Clinicopathological Analysis of 11 Cases]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1099-1106. [PMID: 39507957 PMCID: PMC11536229 DOI: 10.12182/20240960101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Indexed: 11/08/2024]
Abstract
Objective To investigate the clinicopathological features, immunophenotypes, molecular genetic alterations, and prognosis of succinate dehydrogenase-deficient renal cell carcinoma (SDH-RCC). Methods A total of 11 cases of SDH-RCC diagnosed at West China Hospital, Sichuan University between 2016 and 2023 were selected for clinicopathological, immunohistochemical, and DNA sequencing analyses. Results Among the 11 cases of SDH-RCC, there were 5 male patients and 6 female patients. The patients' ages ranged from 12 to 71 years, with an average age of 39.7 years. Among them, 5 patients had tumors located in the right kidney, 5 had tumors located in the left kidney, and 1 patient had bilateral tumors. Microscopic observation showed that the tumor cells of the SDH-RCC patients displayed a wide spectrum of structures, forming sheet-like, nested, and glandular structures. In addition, tumor cells in papillary structures were observed in some cases. The tumor cells had abundant cytoplasm, was eosinophilic, and contained flocculent materials. Intracytoplasmic vacuolations were observed in some of the cells. Among all the patients, 7 (7/11, 63.6%) showed typical low-grade features (grade 1-2 according to the International Society of Urological Pathology [ISUP]/WHO 2016 classification), and 4 (4/11, 36.4%) showed high-grade features (grade 3 according to the ISUP/WHO 2016 classification). The average ages of patients with low-grade and high-grade features were 32.1 years and 58.0 years, respectively. Immunohistochemical staining of all 11 cases demonstrated negative results for SDHB and cytokeratin 7 (CK7), and positive staining results for paired box 8 (PAX-8), fumarate hydratase (FH), and epithelial membrane antigen (EMA). Their Ki-67 index was 1%-30%. In one case, the loss of SDHB expression was also accompanied by a loss of SDHA expression. Sanger sequencing was performed to examine all the exons of SDHB in 7 cases. One case showed a frameshift mutation, c.236Tdel (p.K80Rfs*), and another case harbored a missense mutation, c.725G>A (p.Arg242His). In another case, next generation sequencing revealed that large fragments of SDHB (Exon 4-8 del) were missing. Follow-up data were available for 10 patients. The follow-up time ranged from 4 to 138 months, with the average being 32.8 months, and all patients survived. Metastasis and recurrence were reported in 5 cases, with 3 of them showing high-grade features and 2 showing low-grade features. Conclusion SDH-RCC is rare and the patients demonstrate a relatively young age of onsets. Patients may present with bilateral tumors. Tumors with low-grade features usually occur in young patients, with their Ki-67 index usually being lower than 5%. Individual cases may experience tumor recurrence and metastasis over a long period of follow-up. Tumors with high-grade features tend to occur in older patients who have a higher Ki-67 index, and who are prone to recurrence and metastasis. Negative immunohistochemical staining results for SDHB can assist in tumor diagnosis, but the loss of SDHB protein expression does not necessarily lead to the detection of SDHB gene mutation.
Collapse
Affiliation(s)
- 秀懿 潘
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 昱燕 卫
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 晓晨 隋
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 晓雪 尹
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 林茂 郑
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 浩 曾
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 桥 周
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 铌 陈
- 四川大学华西医院 病理科 (成都 610041)Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Li CY, Jiang HF, Li L, Lai XJ, Liu QR, Yu SB, Yi CL, Chen XQ. Neuroglobin Facilitates Neuronal Oxygenation through Tropic Migration under Hypoxia or Anemia in Rat: How Does the Brain Breathe? Neurosci Bull 2023; 39:1481-1496. [PMID: 36884214 PMCID: PMC10533768 DOI: 10.1007/s12264-023-01040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/03/2023] [Indexed: 03/09/2023] Open
Abstract
The discovery of neuroglobin (Ngb), a brain- or neuron-specific member of the hemoglobin family, has revolutionized our understanding of brain oxygen metabolism. Currently, how Ngb plays such a role remains far from clear. Here, we report a novel mechanism by which Ngb might facilitate neuronal oxygenation upon hypoxia or anemia. We found that Ngb was present in, co-localized to, and co-migrated with mitochondria in the cell body and neurites of neurons. Hypoxia induced a sudden and prominent migration of Ngb towards the cytoplasmic membrane (CM) or cell surface in living neurons, and this was accompanied by the mitochondria. In vivo, hypotonic and anemic hypoxia induced a reversible Ngb migration toward the CM in cerebral cortical neurons in rat brains but did not alter the expression level of Ngb or its cytoplasm/mitochondria ratio. Knock-down of Ngb by RNA interference significantly diminished respiratory succinate dehydrogenase (SDH) and ATPase activity in neuronal N2a cells. Over-expression of Ngb enhanced SDH activity in N2a cells upon hypoxia. Mutation of Ngb at its oxygen-binding site (His64) significantly increased SDH activity and reduced ATPase activity in N2a cells. Taken together, Ngb was physically and functionally linked to mitochondria. In response to an insufficient oxygen supply, Ngb migrated towards the source of oxygen to facilitate neuronal oxygenation. This novel mechanism of neuronal respiration provides new insights into the understanding and treatment of neurological diseases such as stroke and Alzheimer's disease and diseases that cause hypoxia in the brain such as anemia.
Collapse
Affiliation(s)
- Chun-Yang Li
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hai-Feng Jiang
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Li
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Jing Lai
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qian-Rong Liu
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shang-Bin Yu
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cheng-La Yi
- Department of Traumatic Surgery, Tong-ji Hospital, Tong-ji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiao-Qian Chen
- Department of Pathophysiology, Tongji Medical College; Key Laboratory of Neurological Diseases, The Ministry of Education (HUST), Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
7
|
Abou-Rjeileh U, Dos Santos Neto JM, Chirivi M, O'Boyle N, Salcedo D, Prom C, Laguna J, Parales-Giron J, Lock AL, Contreras GA. Oleic acid abomasal infusion limits lipolysis and improves insulin sensitivity in adipose tissue from periparturient dairy cows. J Dairy Sci 2023; 106:4306-4323. [PMID: 37105874 DOI: 10.3168/jds.2022-22402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/03/2023] [Indexed: 04/29/2023]
Abstract
Excessive adipose tissue (AT) lipolysis around parturition in dairy cows is associated with impaired AT insulin sensitivity and increased incidence of metabolic diseases. Supplementing cows with oleic acid (OA) reduces circulating biomarkers of lipolysis and improves energy balance. Nevertheless, it is unclear if OA alters lipid trafficking in AT. In the liver and skeletal muscle, OA improves mitochondrial function and promotes lipid droplet formation by activating perilipin 5 (PLIN5) and peroxisome proliferator-activated receptor α (PPARα). However, it is unknown if this mechanism occurs in AT. The objective of this study was to determine the effect of OA on AT lipolysis, systemic and AT insulin sensitivity, and AT mitochondrial function in periparturient dairy cows. Twelve rumen-cannulated Holstein cows were infused abomasally following parturition with ethanol (CON) or OA (60 g/d) for 14 d. Subcutaneous AT samples were collected at 11 ± 3.6 d before calving (-12 d), and 6 ± 1.0 d (7 d) and 13 ± 1.4 d (14 d) after parturition. An intravenous glucose tolerance test was performed on d 14. Adipocyte morphometry was performed on hematoxylin and eosin-stained AT sections. The antilipolytic effect of insulin (1 μg/L) was evaluated using an ex vivo explant culture following lipolysis stimulation. PLIN5 and PPARα transcription and translation were determined by real-time quantitative PCR and capillary electrophoresis, respectively. RNA sequencing was used to evaluate the transcriptomic profile of mitochondrial gene networks. In CON cows, postpartum lipolysis increased the percentage of smaller (<3,000 µm2) adipocytes at 14 d compared with -12 d. However, OA limited adipocyte size reduction at 14 d. Likewise, OA decreased lipolysis plasma markers nonesterified free fatty acids and β-hydroxybutyrate at 5 and 7 d. Over the 14-d period, compared with CON, OA increased the concentration of plasma insulin and decreased plasma glucose. During the glucose tolerance test, OA decreased circulating glucose concentration (at 10, 20, 30, 40 min) and the glucose clearance rate. Moreover, OA increased insulin at 10 and 20 min and tended to increase it at 30 min. Following lipolysis stimulation, OA improved the antilipolytic effect of insulin in the AT at 14 d. PLIN5 and PPARA gene expression decreased postpartum regardless of treatment. However, OA increased PLIN5 protein expression at 14 d and increased PPARA at 7 and 14 d. Immunohistochemical analysis of AT and RNA sequencing data showed that OA increased the number of mitochondria and improved mitochondrial function. However, OA had no effect on production and digestibility. Our results demonstrate that OA limits AT lipolysis, improves systemic and AT insulin sensitivity, and is associated with markers of mitochondrial function supporting a shift to lipogenesis in AT of periparturient dairy cows.
Collapse
Affiliation(s)
- Ursula Abou-Rjeileh
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - José M Dos Santos Neto
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing 48824
| | - Miguel Chirivi
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - Nial O'Boyle
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, United Kingdom
| | - David Salcedo
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - Crystal Prom
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing 48824
| | - Juliana Laguna
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - Jair Parales-Giron
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing 48824
| | - Adam L Lock
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing 48824.
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824.
| |
Collapse
|
8
|
Alfatni A, Charles AL, Sauer F, Riou M, Goupilleau F, Talha S, Meyer A, Andres E, Kindo M, Mazzucotelli JP, Epailly E, Geny B. Peripheral Blood Mononuclear Cells Mitochondrial Respiration and Superoxide Anion after Heart Transplantation. J Clin Med 2022; 11:jcm11237247. [PMID: 36498821 PMCID: PMC9735976 DOI: 10.3390/jcm11237247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The mitochondrial function of circulating peripheral blood mononuclear cells (PBMCs) is an interesting new approach to cardiac diseases. Thus, PBMC's mitochondrial respiration decreases in relation to heart failure severity. However, no data are available on heart-transplanted patients (Htx). POPULATION AND METHODS We determined PBMCs mitochondrial respiration by high-resolution respirometry (Oroboros Instruments) and superoxide anion production using electron paramagnetic resonance (Bruker-Biospin) in 20 healthy subjects and 20 matched Htx and investigated clinical, biological, echocardiographic, coronarography and biopsy characteristics. RESULTS PBMCs mitochondrial respiratory chain complex II respiration was decreased in Htx (4.69 ± 0.84 vs. 7.69 ± 1.00 pmol/s/million cell in controls and Htx patients, respectively; p = 0.007) and complex IV respiration was increased (24.58 ± 2.57 vs. 15.68 ± 1.67 pmol/s/million cell; p = 0.0035). Superoxide anion production was also increased in Htx (1.47 ± 0.10 vs. 1.15 ± 0.10 µmol/min; p = 0.041). The leucocyte-to-lymphocyte ratio was increased in Htx, whom complex II correlated with leucocyte number (r = 0.51, p = 0.02) and with the left ventricular posterior wall peak early diastolic myocardial velocity (r = -0.62, p = 0.005). Complex IV was increased in the two patients with acute rejection and correlated negatively with Htx's isovolumetric relation time (r = -0.45, p = 0.045). DISCUSSION Although presenting with normal systolic function, Htx demonstrated abnormal PBMC's mitochondrial respiration. Unlike immunosuppressive therapies, subclinical diastolic dysfunction might be involved in these changes. Additionally, lymphopenia might reduce complex II, and acute rejection enhances complex IV respirations. CONCLUSION PBMC's mitochondrial respiration appears modified in Htx, potentially linked to cellular shift, mild diastolic dysfunction and/or acute rejection.
Collapse
Affiliation(s)
- Abrar Alfatni
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
| | - Anne-Laure Charles
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
| | - François Sauer
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Marianne Riou
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Fabienne Goupilleau
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
| | - Samy Talha
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Alain Meyer
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Emmanuel Andres
- Department of Internal Medicine, University Hospital of Strasbourg, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Michel Kindo
- Cardiovascular Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Jean-Philippe Mazzucotelli
- Cardiovascular Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Eric Epailly
- Cardiovascular Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
| | - Bernard Geny
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 11 Rue Humann, 67000 Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, NHC, 1 Place de l’Hôpital, CEDEX, 67091 Strasbourg, France
- Correspondence:
| |
Collapse
|
9
|
Wang L, Cybula M, Rostworowska M, Wang L, Mucha P, Bulicz M, Bieniasz M. Upregulation of Succinate Dehydrogenase (SDHA) Contributes to Enhanced Bioenergetics of Ovarian Cancer Cells and Higher Sensitivity to Anti-Metabolic Agent Shikonin. Cancers (Basel) 2022; 14:5097. [PMID: 36291881 PMCID: PMC9599980 DOI: 10.3390/cancers14205097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022] Open
Abstract
We discovered that the overexpression of mitochondrial enzyme succinate dehydrogenase (SDHA) is particularly prevalent in ovarian carcinoma and promotes highly metabolically active phenotype. Succinate dehydrogenase deficiency has been previously studied in some rare disorders. However, the role of SDHA upregulation and its impact on ovarian cancer metabolism has never been investigated, emphasizing the need for further research. We investigated the functional consequences of SDHA overexpression in ovarian cancer. Using proteomics approaches and biological assays, we interrogated protein content of metabolic pathways, cell proliferation, anchorage-independent growth, mitochondrial respiration, glycolytic function, and ATP production rates in those cells. Lastly, we performed a drug screening to identify agents specifically targeting the SDHA overexpressing tumor cells. We showed that SDHA overexpressing cells are characterized by enhanced energy metabolism, relying on both glycolysis and oxidative phosphorylation to meet their energy needs. In addition, SDHA-high phenotype was associated with cell vulnerability to glucose and glutamine deprivation, which led to a substantial reduction of ATP yield. We also identified an anti-metabolic compound shikonin with a potent efficacy against SDHA overexpressing ovarian cancer cells. Our data underline the unappreciated role of SDHA in reprogramming of ovarian cancer metabolism, which represents a new opportunity for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Magdalena Bieniasz
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
10
|
PIKE-A Modulates Mitochondrial Metabolism through Increasing SDHA Expression Mediated by STAT3/FTO Axis. Int J Mol Sci 2022; 23:ijms231911304. [PMID: 36232604 PMCID: PMC9570435 DOI: 10.3390/ijms231911304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Previous studies have shown that phosphoinositide 3-kinase enhancer-activating Akt (PIKE-A) is involved in the regulation of several biological processes in cancer. In our previous study, we demonstrated a crucial function of PIKE-A in cancer energy metabolism by regulating pentose phosphate pathway (PPP) flux. However, whether PIKE-A regulates energy metabolism through affecting mitochondrial changes are poorly understood. In the present study, we show that PIKE-A promotes mitochondrial membrane potential, leading to increasing proliferation of glioblastoma cell. Mechanistically, PIKE-A affects the expression of respiratory chain complex Ⅱ succinate dehydrogenase A (SDHA), mediated by regulating the axis of STAT3/FTO. Taken together, these results revealed that inhibition of PIKE-A reduced STAT3/FTO/SDHA expression, leading to the suppression of mitochondrial function. Thus, our findings suggest the PIKE-A/STAT3/FTO/SDHA axis as promising anti-cancer treatment targets.
Collapse
|
11
|
Bénit P, Goncalves J, El Khoury R, Rak M, Favier J, Gimenez-Roqueplo AP, Rustin P. Succinate Dehydrogenase, Succinate, and Superoxides: A Genetic, Epigenetic, Metabolic, Environmental Explosive Crossroad. Biomedicines 2022; 10:1788. [PMID: 35892689 PMCID: PMC9394281 DOI: 10.3390/biomedicines10081788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Research focused on succinate dehydrogenase (SDH) and its substrate, succinate, culminated in the 1950s accompanying the rapid development of research dedicated to bioenergetics and intermediary metabolism. This allowed researchers to uncover the implication of SDH in both the mitochondrial respiratory chain and the Krebs cycle. Nowadays, this theme is experiencing a real revival following the discovery of the role of SDH and succinate in a subset of tumors and cancers in humans. The aim of this review is to enlighten the many questions yet unanswered, ranging from fundamental to clinically oriented aspects, up to the danger of the current use of SDH as a target for a subclass of pesticides.
Collapse
Affiliation(s)
- Paule Bénit
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Goncalves
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Riyad El Khoury
- Department of Pathology and Laboratory Medicine, Neuromuscular Diagnostic Laboratory, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Malgorzata Rak
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Favier
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Anne-Paule Gimenez-Roqueplo
- Département de Médecine Génomique des Tumeurs et des Cancers, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, F-75015 Paris, France;
| | - Pierre Rustin
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| |
Collapse
|
12
|
Cardiac disruption of SDHAF4-mediated mitochondrial complex II assembly promotes dilated cardiomyopathy. Nat Commun 2022; 13:3947. [PMID: 35803927 PMCID: PMC9270418 DOI: 10.1038/s41467-022-31548-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
Succinate dehydrogenase, which is known as mitochondrial complex II, has proven to be a fascinating machinery, attracting renewed and increased interest in its involvement in human diseases. Herein, we find that succinate dehydrogenase assembly factor 4 (SDHAF4) is downregulated in cardiac muscle in response to pathological stresses and in diseased hearts from human patients. Cardiac loss of Sdhaf4 suppresses complex II assembly and results in subunit degradation and complex II deficiency in fetal mice. These defects are exacerbated in young adults with globally impaired metabolic capacity and activation of dynamin-related protein 1, which induces excess mitochondrial fission and mitophagy, thereby causing progressive dilated cardiomyopathy and lethal heart failure in animals. Targeting mitochondria via supplementation with fumarate or inhibiting mitochondrial fission improves mitochondrial dynamics, partially restores cardiac function and prolongs the lifespan of mutant mice. Moreover, the addition of fumarate is found to dramatically improve cardiac function in myocardial infarction mice. These findings reveal a vital role for complex II assembly in the development of dilated cardiomyopathy and provide additional insights into therapeutic interventions for heart diseases. Functional succinate dehydrogenase (SDH) complex is vital to mitochondrial homeostasis. Here the authors show that disruption of SDH assembly in the heart causes dilated cardiomyopathy via impairing the mitochondrial integrity and metabolism and that mitochondrial interventions can be an effective approach to ameliorate the disease progression.
Collapse
|
13
|
Abstract
Although tumourigenesis occurs due to genetic mutations, the role of epigenetic dysregulations in cancer is also well established. Epigenetic dysregulations in cancer may occur as a result of mutations in genes encoding histone/DNA-modifying enzymes and chromatin remodellers or mutations in histone protein itself. It is also true that misregulated gene expression without genetic mutations in these factors could also support tumour initiation and progression. Interestingly, metabolic rewiring has emerged as a hallmark of cancer due to gene mutations in specific metabolic enzymes or dietary/environmental factors. Recent studies report an intricate cross-talk between epigenetic and metabolic reprogramming in cancer. This review discusses the role of epigenetic and metabolic dysregulations and their cross-talk in tumourigenesis with a special focus on gliomagenesis. We also discuss the role of recently developed human embryonic stem cells/induced pluripotent stem cells-derived organoid models of gliomas and how these models are proving instrumental in uncovering human-specific cellular and molecular complexities of gliomagenesis.
Collapse
Affiliation(s)
- Bismi Phasaludeen
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
14
|
Lamy C, Mansard C, Blondel L, Mercier L, Paci A, Broutin S. Quantification of succinic acid levels, linked to succinate dehydrogenase (SDH) dysfunctions, by an automated and fully validated liquid chromatography tandem mass spectrometry method suitable for multi-matrix applications. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1189:123085. [PMID: 34974318 DOI: 10.1016/j.jchromb.2021.123085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/06/2021] [Accepted: 12/12/2021] [Indexed: 11/24/2022]
Abstract
The hallmarks of cancer include metabolism with deregulating cellular energetics. Dysfunctions in succinate dehydrogenase (SDH) metabolic enzyme activity, leading to an abnormal accumulation of succinic acid has been described in solid tumors but also in inflammation and ischemia reperfusion injury. Succinic acid is a potential biomarker of SDH related pathologies for diagnostic, evaluation of treatment response and follow-up of the disease. We developed a liquid chromatography tandem mass spectrometry (LC-MS/MS) method allowing a rapid, accurate and precise quantification of succinic acid levels in clinical (serum, urine) and preclinical (cellular pellets, supernatants) samples. 13C4 succinic acid disodium salt was used as internal standard and added to samples before a solid phase extraction (SPE) on Phenomenex STRATATM XL-A (200 mg - 3 mL) 33 µm cartridges. This method is automated by a Freedom EVO® platform from TECAN and succinic acid is separated on a C18 column combined to a Xevo® TQ-S micro Waters mass spectrometer with electrospray ionization (ESI) source. This biomedical analysis allows standard curves to be linear over the range 1.0-135.5 µM with r2 values > 0.999 and low matrix effects (<9.1 %). This method, which is validated according updated European Medicine Agency (EMA) guidelines, is accurate between-run (<11.0 %) and within-run (<7.8 %), precise between-run (<14.4 CV %) and within-run (<3.7 CV %), and is suitable for clinical and preclinical applications.
Collapse
Affiliation(s)
- Constance Lamy
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, INSERM1030, Radiothérapie moléculaire et innovation thérapeutique, F-94805 Villejuif, France
| | - Clémence Mansard
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France
| | - Louis Blondel
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France
| | - Lionel Mercier
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France
| | - Angelo Paci
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, INSERM1030, Radiothérapie moléculaire et innovation thérapeutique, F-94805 Villejuif, France; Service de pharmacocinétique, Faculté de pharmacie, Université Paris-Saclay, F-92296 Châtenay-Malabry, France
| | - Sophie Broutin
- Gustave Roussy, Service de Pharmacologie, Département de biologie et pathologie médicales, F-94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, INSERM1030, Radiothérapie moléculaire et innovation thérapeutique, F-94805 Villejuif, France.
| |
Collapse
|
15
|
SDHI Fungicide Toxicity and Associated Adverse Outcome Pathways: What Can Zebrafish Tell Us? Int J Mol Sci 2021; 22:ijms222212362. [PMID: 34830252 PMCID: PMC8618699 DOI: 10.3390/ijms222212362] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/19/2023] Open
Abstract
Succinate dehydrogenase inhibitor (SDHI) fungicides are increasingly used in agriculture to combat molds and fungi, two major threats to both food supply and public health. However, the essential requirement for the succinate dehydrogenase (SDH) complex—the molecular target of SDHIs—in energy metabolism for almost all extant eukaryotes and the lack of species specificity of these fungicides raise concerns about their toxicity toward off-target organisms and, more generally, toward the environment. Herein we review the current knowledge on the toxicity toward zebrafish (Brachydanio rerio) of nine commonly used SDHI fungicides: bixafen, boscalid, fluxapyroxad, flutolanil, isoflucypram, isopyrazam, penthiopyrad, sedaxane, and thifluzamide. The results indicate that these SDHIs cause multiple adverse effects in embryos, larvae/juveniles, and/or adults, sometimes at developmentally relevant concentrations. Adverse effects include developmental toxicity, cardiovascular abnormalities, liver and kidney damage, oxidative stress, energy deficits, changes in metabolism, microcephaly, axon growth defects, apoptosis, and transcriptome changes, suggesting that glycometabolism deficit, oxidative stress, and apoptosis are critical in the toxicity of most of these SDHIs. However, other adverse outcome pathways, possibly involving unsuspected molecular targets, are also suggested. Lastly, we note that because of their recent arrival on the market, the number of studies addressing the toxicity of these compounds is still scant, emphasizing the need to further investigate the toxicity of all SDHIs currently used and to identify their adverse effects and associated modes of action, both alone and in combination with other pesticides.
Collapse
|
16
|
Sturrock BRH, Macnamara EF, McGuire P, Kruk S, Yang I, Murphy J, Tifft CJ, Gordon‐Lipkin E. Progressive cerebellar atrophy in a patient with complex II and III deficiency and a novel deleterious variant in SDHA: A Counseling Conundrum. Mol Genet Genomic Med 2021; 9:e1692. [PMID: 33960148 PMCID: PMC8222855 DOI: 10.1002/mgg3.1692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Complex II is an essential component of the electron transport chain, linking it with the tricarboxylic acid cycle. Its four subunits are encoded in the nuclear genome, and deleterious variants in these genes, including SDHA (OMIM 600857), are associated with a wide range of symptoms including neurological disease, cardiomyopathy, and neoplasia (paraganglioma-pheochromocytomas (PGL/PCC), and gastrointestinal stromal tumors). Deleterious variants of SDHA are most frequently associated with Leigh and Leigh-like syndromes. METHODS AND RESULTS Here, we describe a case of a 9-year-old boy with tremor, nystagmus, hypotonia, developmental delay, significant ataxia, and progressive cerebellar atrophy. He was found to have biallelic variants in SDHA, a known pathogenic variant (c.91C>T (p.R31*)), and a variant of unknown significance (c.454G>A (p.E152K)). Deficient activity of complexes II and III was detected in fibroblasts from the patient consistent with a diagnosis of a respiratory chain disorder. CONCLUSION We, therefore, consider whether c.454G>A (p.E152K) is, indeed, a pathogenic variant, and what implications it has for family members who carry the same variant.
Collapse
Affiliation(s)
- Beattie R. H. Sturrock
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
- Brighton and Sussex University Hospitals NHS TrustBrightonEngland
| | - Ellen F. Macnamara
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
| | - Peter McGuire
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Shannon Kruk
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Ivan Yang
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Jennifer Murphy
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
| | - Cyndi J. Tifft
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
- Office of the Clinical DirectorNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Eliza Gordon‐Lipkin
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| |
Collapse
|
17
|
Godel M, Ortone G, Anobile DP, Pasino M, Randazzo G, Riganti C, Kopecka J. Targeting Mitochondrial Oncometabolites: A New Approach to Overcome Drug Resistance in Cancer. Pharmaceutics 2021; 13:762. [PMID: 34065551 PMCID: PMC8161136 DOI: 10.3390/pharmaceutics13050762] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/28/2022] Open
Abstract
Drug resistance is the main obstacle for a successful cancer therapy. There are many mechanisms by which cancers avoid drug-mediated death, including alterations in cellular metabolism and apoptotic programs. Mitochondria represent the cell's powerhouse and the connection between carbohydrate, lipid and proteins metabolism, as well as crucial controllers of apoptosis, playing an important role not only in tumor growth and progression, but also in drug response. Alterations in tricarboxylic acid cycle (TCA) caused by mutations in three TCA enzymes-isocitrate dehydrogenase, succinate dehydrogenase and fumarate hydratase-lead to the accumulation of 2-hydroxyglutarate, succinate and fumarate respectively, collectively known as oncometabolites. Oncometabolites have pleiotropic effects on cancer biology. For instance, they generate a pseudohypoxic phenotype and induce epigenetic changes, two factors that may promote cancer drug resistance leading to disease progression and poor therapy outcome. This review sums up the most recent findings about the role of TCA-derived oncometabolites in cancer aggressiveness and drug resistance, highlighting possible pharmacological strategies targeting oncometabolites production in order to improve the efficacy of cancer treatment.
Collapse
|
18
|
Sulaimon LA, Adisa RA, Samuel TA, Joel IY, Ayankojo AG, Abdulkareem FB, Olaniyi TO. Molecular mechanism of mitoquinol mesylate in mitigating the progression of hepatocellular carcinoma-in silico and in vivo studies. J Cell Biochem 2021; 122:1157-1172. [PMID: 33909925 DOI: 10.1002/jcb.29937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The safety and efficacy of mitoquinol mesylate (MitoQ) in attenuating the progression of hepatocellular carcinoma (HCC) in Wistar rats has been reported. However, the binding modes for MitoQ as well as its molecular mechanisms in cirrhosis and liver cancer have not been fully investigated. This study sought to understand the structural and molecular mechanisms of MitoQ in modulating the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and mitochondrial succinate dehydrogenase (SDH) in cirrhotic-HCC rats. The research indicates that the upregulated Nrf2 expression in cirrhotic-HCC rats was significantly (p < 0.05) reduced by MitoQ while the activity of SDH was significantly (p < 0.05) increased. Analysis of binding modes revealed MitoQ interacts with amino acid residues in the active pocket of tramtrack and bric-a-brac (BTB) and KELCH domains of KEAP1 with average binding affinities of -66.46 and -74.74 kcal/mol, respectively. Also, MitoQ interacted with the key amino acid residues at the active site of mitochondrial complex II with a higher average binding affinity of -75.76 kcal/mol compared to co-crystallized ligand of complex II (-62.31 kcal/mol). Molecular dynamics simulations data showed the binding of MitoQ to be stable with low eigenvalues while the quantum mechanics calculations suggest MitoQ to be very reactive with its mechanism of chemical reactivity to be via electrophilic reactions. Thus, MitoQ modulates expression of Nrf2 and enhances activity of mitochondrial SDH in cirrhotic-HCC rats via its interaction with key amino acid residues in the active pocket of BTB and KELCH domains of KEAP1 as well as amino residues at the active site of SDH. These findings are significant in demonstrating the potential of Nrf2 and SDH as possible biomarkers for the diagnosis and/or prognosis of hepatocellular carcinoma in patients. This study also supports repurposing of mitoQ for the treatment/management of liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Lateef Adegboyega Sulaimon
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Lagos, Nigeria
| | - Rahmat Adetutu Adisa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Lagos, Nigeria
| | - Titilola Aderonke Samuel
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Lagos, Nigeria
| | | | - Akinrinade George Ayankojo
- Department of Materials and Environmental Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Fatimah Biade Abdulkareem
- Department of Anatomic and Molecular Pathology, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Lagos, Nigeria
| | - Timothy Olajire Olaniyi
- Department of Science Laboratory Technology, Faculty of Science, Oyo State College of Agriculture and Technology, Igbo-ora, Oyo, Nigeria
| |
Collapse
|
19
|
Fernandez-Vizarra E, Zeviani M. Mitochondrial disorders of the OXPHOS system. FEBS Lett 2020; 595:1062-1106. [PMID: 33159691 DOI: 10.1002/1873-3468.13995] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/21/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
Mitochondrial disorders are among the most frequent inborn errors of metabolism, their primary cause being the dysfunction of the oxidative phosphorylation system (OXPHOS). OXPHOS is composed of the electron transport chain (ETC), formed by four multimeric enzymes and two mobile electron carriers, plus an ATP synthase [also called complex V (cV)]. The ETC performs the redox reactions involved in cellular respiration while generating the proton motive force used by cV to synthesize ATP. OXPHOS biogenesis involves multiple steps, starting from the expression of genes encoded in physically separated genomes, namely the mitochondrial and nuclear DNA, to the coordinated assembly of components and cofactors building each individual complex and eventually the supercomplexes. The genetic cause underlying around half of the diagnosed mitochondrial disease cases is currently known. Many of these cases result from pathogenic variants in genes encoding structural subunits or additional factors directly involved in the assembly of the ETC complexes. Here, we review the historical and most recent findings concerning the clinical phenotypes and the molecular pathological mechanisms underlying this particular group of disorders.
Collapse
Affiliation(s)
- Erika Fernandez-Vizarra
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Massimo Zeviani
- Venetian Institute of Molecular Medicine, Padova, Italy.,Department of Neurosciences, University of Padova, Italy
| |
Collapse
|
20
|
De Filpo G, Cilotti A, Rolli L, Pastorino U, Sonzogni A, Pradella S, Cantini G, Ercolino T, Nesi G, Mannelli M, Maggi M, Canu L. SDHx and Non-Chromaffin Tumors: A Mediastinal Germ Cell Tumor Occurring in a Young Man with Germline SDHB Mutation. ACTA ACUST UNITED AC 2020; 56:medicina56110561. [PMID: 33113876 PMCID: PMC7693473 DOI: 10.3390/medicina56110561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/31/2022]
Abstract
Background: Mutations in genes encoding one of the subunits of succinate dehydrogenase (SDH) are involved in pheochromocytoma (PHEO) and paraganglioma (PGL) development. Over the last few years, such mutations have also been associated with non-chromaffin tumors. However, immunohistochemistry (IHC) on the tumor tissue and a study on the loss of heterozygosity (LOH) aimed at demonstrating the pathogenic role of SDHx genes have only been employed in a few cases. Case report: We describe the case of a 19-year-old Caucasian man with a germline SDHB mutation, who presented with acne vulgaris resistant to medical treatment. His follow-up for chromaffin tumors was negative, while hormonal tests revealed suppressed gonadotropins with testosterone in the upper range of normality and elevated β-human chorionic gonadotropin (β-hCG). At the whole-body enhanced CT scan, a mediastinal lesion suggestive of a germ cell tumor (GCT) was detected. 18FDG-PET (fluorodeoxyglucose-positron emission tomography) imaging showed low glucose metabolism at the mediastinal site. Surgical removal of the mass was uneventful. Pathology confirmed the diagnosis of GCT consisting of cystic teratoma (95%) and seminoma (5%). IHC for SDHB showed normal protein expression, and genetic analysis of the tumor tissue revealed the absence of SDHB LOH. Normalization of the hormonal tests and acne attenuation were achieved after surgery. Conclusion: We report an incidental association of a germinal SDHB mutation and mediastinal GCT in a young Caucasian man. Our paper highlights the importance of IHC and genetic analysis in confirming the etiologic role of SDHx genes in nonchromaffin tumors, thus excluding incidental associations.
Collapse
Affiliation(s)
- Giuseppina De Filpo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (G.C.); (M.M.); (M.M.); (L.C.)
- Endocrinology Unit, Careggi University Hospital, 50139 Florence, Italy; (A.C.); (T.E.)
- Correspondence: ; Tel.: +39-55-2758241
| | - Antonio Cilotti
- Endocrinology Unit, Careggi University Hospital, 50139 Florence, Italy; (A.C.); (T.E.)
| | - Luigi Rolli
- Division of Thoracic Surgery, IRCCS Foundation, Istituto Nazionale dei Tumori, 20133 Milan, Italy; (L.R.); (U.P.)
| | - Ugo Pastorino
- Division of Thoracic Surgery, IRCCS Foundation, Istituto Nazionale dei Tumori, 20133 Milan, Italy; (L.R.); (U.P.)
| | - Angelica Sonzogni
- Department of Pathology and Laboratory Medicine, IRCCS Foundation, Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Silvia Pradella
- Department of Radiology, Careggi University Hospital, 50139 Florence, Italy;
| | - Giulia Cantini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (G.C.); (M.M.); (M.M.); (L.C.)
| | - Tonino Ercolino
- Endocrinology Unit, Careggi University Hospital, 50139 Florence, Italy; (A.C.); (T.E.)
| | - Gabriella Nesi
- Department of Health Sciences, Division of Pathological Anatomy, University of Florence, 50139 Florence, Italy;
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (G.C.); (M.M.); (M.M.); (L.C.)
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (G.C.); (M.M.); (M.M.); (L.C.)
- Endocrinology Unit, Careggi University Hospital, 50139 Florence, Italy; (A.C.); (T.E.)
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (G.C.); (M.M.); (M.M.); (L.C.)
- Endocrinology Unit, Careggi University Hospital, 50139 Florence, Italy; (A.C.); (T.E.)
| |
Collapse
|
21
|
Immunohistochemistry and Mutation Analysis of SDHx Genes in Carotid Paragangliomas. Int J Mol Sci 2020; 21:ijms21186950. [PMID: 32971818 PMCID: PMC7576476 DOI: 10.3390/ijms21186950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Carotid paragangliomas (CPGLs) are rare neuroendocrine tumors often associated with mutations in SDHx genes. The immunohistochemistry of succinate dehydrogenase (SDH) subunits has been considered a useful instrument for the prediction of SDHx mutations in paragangliomas/pheochromocytomas. We compared the mutation status of SDHx genes with the immunohistochemical (IHC) staining of SDH subunits in CPGLs. To identify pathogenic/likely pathogenic variants in SDHx genes, exome sequencing data analysis among 42 CPGL patients was performed. IHC staining of SDH subunits was carried out for all CPGLs studied. We encountered SDHx variants in 38% (16/42) of the cases in SDHx genes. IHC showed negative (5/15) or weak diffuse (10/15) SDHB staining in most tumors with variants in any of SDHx (94%, 15/16). In SDHA-mutated CPGL, SDHA expression was completely absent and weak diffuse SDHB staining was detected. Positive immunoreactivity for all SDH subunits was found in one case with a variant in SDHD. Notably, CPGL samples without variants in SDHx also demonstrated negative (2/11) or weak diffuse (9/11) SDHB staining (42%, 11/26). Obtained results indicate that SDH immunohistochemistry does not fully reflect the presence of mutations in the genes; diagnostic effectiveness of this method was 71%. However, given the high sensitivity of SDHB immunohistochemistry, it could be used for initial identifications of patients potentially carrying SDHx mutations for recommendation of genetic testing.
Collapse
|
22
|
Wai Hon K, Zainal Abidin SA, Othman I, Naidu R. Insights into the Role of microRNAs in Colorectal Cancer (CRC) Metabolism. Cancers (Basel) 2020; 12:cancers12092462. [PMID: 32878019 PMCID: PMC7565715 DOI: 10.3390/cancers12092462] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers, with a high mortality rate globally. The pathophysiology of CRC is mainly initiated by alteration in gene expression, leading to dysregulation in multiple signalling pathways and cellular processes. Metabolic reprogramming is one of the important cancer hallmarks in CRC, which involves the adaptive changes in tumour cell metabolism to sustain the high energy requirements for rapid cell proliferation. There are several mechanisms in the metabolic reprogramming of cancer cells, such as aerobic glycolysis, oxidative phosphorylation, lactate and fatty acids metabolism. MicroRNAs (miRNAs) are a class of non-coding RNAs that are responsible for post-transcriptional regulation of gene expression. Differential expression of miRNAs has been shown to play an important role in different aspects of tumorigenesis, such as proliferation, apoptosis, and drug resistance, as well as metabolic reprogramming. Increasing evidence also reports that miRNAs could function as potential regulators of metabolic reprogramming in CRC cells. This review provides an insight into the role of different miRNAs in regulating the metabolism of CRC cells as well as to discuss the potential role of miRNAs as biomarkers or therapeutic targets in CRC tumour metabolism.
Collapse
|
23
|
Next-generation sequencing identifies recurrent copy number variations in invasive breast carcinomas from Ghana. Mod Pathol 2020; 33:1537-1545. [PMID: 32152520 PMCID: PMC7390688 DOI: 10.1038/s41379-020-0515-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 01/31/2023]
Abstract
African and African-American (AA) women have higher incidence of triple-negative breast cancers (TNBC) with high histological grade and aggressive clinical behavior, but the reasons are not fully understood. We recently found that the oncogenic protein EZH2 is overexpressed in Ghanaian breast cancer patients, with 16% of the tumors expressing cytoplasmic EZH2. Understanding the molecular underpinnings of these aggressive tumors may lead to the identification of potential targetable oncogenic drivers. We characterized the copy number variations of 11 Ghanaian breast tumor patients by targeted multiplexed PCR-based DNA next-generation sequencing (NGS) over 130 cancer-relevant genes. While the DNA quality was not optimal for mutation analysis, 90% of the tumors had frequent recurrent copy number alterations (CNAs) of 17 genes: SDHC, RECQL4, TFE3, BCL11A, BCL2L1, PDGFRA, DEK, SMUG1, AKT3, SMARCA4, VHL, KLF6, CCNE1, G6PD, FGF3, ABL1, and CCND1, with the top oncogenic functions being mitotic G1-G1/S-phase regulation, gene transcription, apoptosis, and PI3K/AKT pathway. The most common recurrent high-level CNAs were gains of RECQL4 and SDHC, in 50% and 60% of cases, respectively. Network analyses revealed a significant predicted interaction among 12 of the 17 (70.6%) genes with high-level CNAs (p = 5.7E-07), which was highly correlated with EZH2 expression (r = 0.4-0.75). By immunohistochemistry, RECQL4 and SDHC proteins were upregulated in 53 of 86 (61.6%) and 48 of 86 (56%) of Ghanaian invasive carcinoma tissue samples. In conclusion, our data show that invasive carcinomas from Ghana exhibit recurrent CNAs in 17 genes, with functions in oncogenic pathways, including PI3K/AKT and G1-G1/S regulation, which may have implications for the biology and treatment of invasive carcinomas in African and AA women.
Collapse
|
24
|
Sun L, Zhou F, Shao Y, Lv Z, Li C. The iron-sulfur protein subunit of succinate dehydrogenase is critical in driving mitochondrial reactive oxygen species generation in Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2020; 102:350-360. [PMID: 32371258 DOI: 10.1016/j.fsi.2020.04.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 06/11/2023]
Abstract
Succinate dehydrogenase (SDH) is a mitochondrial enzyme with the unique ability to participate in both the tricarboxylic acid cycle and the electron transport chain to produce reactive oxygen species (ROS). The B subunit of SDH is required for succinate oxidation, which is critical for pro-inflammatory response. In this study, we cloned the iron-sulfur protein subunit of SDH from Apostichopus japonicus (denoted as AjSDHB) via RACE technology and explored its role in the immune system as a response to pathogen infection. The full-length cDNA of AjSDHB was 1442 bp with a complete open reading frame of 858 bp encoding 286 amino acids. Simple modular architecture research tool analysis revealed that AjSDHB contained two conserved domains, including a 2Fe-2S iron-sulfur cluster binding domain and a 4Fe-4S dicluster domain, without a signal peptide. Multiple sequence alignment demonstrated that AjSDHB shared a high degree of structural conservation and sequence identities with other counterparts from invertebrates and vertebrates. Phylogenetic analysis supported the finding that AjSDHB is a new member of the SDHB protein subfamily. Tissue distribution analysis revealed that AjSDHB was expressed in all examined tissues and particularly highly expressed in the muscles. AjSDHB transcripts were markedly induced in coelomocytes both by Vibrio splendidus challenge in vivo and lipopolysaccharide exposure in vitro. Function analysis showed that siRNA-mediated AjSDHB knockdown could substantially reduce the mitochondrial membrane potential (ΔΨm) and further decrease mitochondrial ROS production in A. japonicus coelomocytes. By contrast, AjSDHB overexpression considerably increased ΔΨm and mitochondrial ROS production of A. japonicus coelomocytes. These results supported the idea that AjSDHB is involved in the innate immunity of A. japonicus through its participation in mitochondrial ROS generation.
Collapse
Affiliation(s)
- Lianlian Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Fangyuan Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Yina Shao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Zhimeng Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
25
|
Marchetti L, Perrucci L, D'Ercole F, Zatelli MC, Ambrosio MR, Giganti M, Carnevale A. Neck paraganglioma and follicular lymphoma: a case report. J Med Case Rep 2019; 13:376. [PMID: 31856921 PMCID: PMC6924013 DOI: 10.1186/s13256-019-2323-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
Background Paragangliomas and pheochromocytomas are sympathetic or parasympathetic tumors derived from the paraganglia and the adrenal medulla, respectively. Paragangliomas and pheochromocytomas can be sporadic or familial, the latter frequently being multifocal and possibly due to succinate dehydrogenase complex genes mutations. In addition, 12% of sporadic paragangliomas are related to covered succinate dehydrogenase complex mutations. The importance of identifying succinate dehydrogenase complex mutations is related to the risk for these patients of developing multiple tumors, including non-endocrine ones, showing an aggressive clinical presentation. Case presentation We report the case of a 45-year-old Caucasian man with an indolent mass in his neck. Ultrasound of his neck, magnetic resonance imaging, and 1,4,7,10-tetraazacyclododecane-N(I),N(II),N(III),N(IIII)-tetraacetic acid(D)-Phe(1)-thy(3)-octreotide (68Ga-DOTATOC) positron emission tomography-computed tomography and endocrine work-up were consistent with a carotid body paraganglioma with concomitant nodal enlargement in several body regions, which turned out to be a follicular lymphoma at histology. He was found to carry a germline Succinate dehydrogenase subunit B gene (SDHB) mutation. Conclusion It is crucial to look for a second malignancy in the case of a paraganglioma demonstrating succinate dehydrogenase complex germline mutations.
Collapse
Affiliation(s)
- Lara Marchetti
- Department of Interventional and Diagnostic Radiology, Arcispedale Sant'Anna, Ferrara, Italy
| | - Luca Perrucci
- Section of Diagnostic Imaging, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy. .,University Radiology Unit, Radiology Department, Sant'Anna University Hospital, Ferrara, Italy.
| | - Francesca D'Ercole
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Maria Rosaria Ambrosio
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Melchiore Giganti
- Section of Diagnostic Imaging, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Aldo Carnevale
- University Radiology Unit, Radiology Department, Sant'Anna University Hospital, Ferrara, Italy
| |
Collapse
|
26
|
Bacolla A, Ye Z, Ahmed Z, Tainer JA. Cancer mutational burden is shaped by G4 DNA, replication stress and mitochondrial dysfunction. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 147:47-61. [PMID: 30880007 PMCID: PMC6745008 DOI: 10.1016/j.pbiomolbio.2019.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 02/01/2023]
Abstract
A hallmark of cancer is genomic instability, which can enable cancer cells to evade therapeutic strategies. Here we employed a computational approach to uncover mechanisms underlying cancer mutational burden by focusing upon relationships between 1) translocation breakpoints and the thousands of G4 DNA-forming sequences within retrotransposons impacting transcription and exemplifying probable non-B DNA structures and 2) transcriptome profiling and cancer mutations. We determined the location and number of G4 DNA-forming sequences in the Genome Reference Consortium Human Build 38 and found a total of 358,605 covering ∼13.4 million bases. By analyzing >97,000 unique translocation breakpoints from the Catalogue Of Somatic Mutations In Cancer (COSMIC), we found that breakpoints are overrepresented at G4 DNA-forming sequences within hominid-specific SVA retrotransposons, and generally occur in tumors with mutations in tumor suppressor genes, such as TP53. Furthermore, correlation analyses between mRNA levels and exome mutational loads from The Cancer Genome Atlas (TCGA) encompassing >450,000 gene-mutation regressions revealed strong positive and negative associations, which depended upon tissue of origin. The strongest positive correlations originated from genes not listed as cancer genes in COSMIC; yet, these show strong predictive power for survival in most tumor types by Kaplan-Meier estimation. Thus, correlation analyses of DNA structure and gene expression with mutation loads complement and extend more traditional approaches to elucidate processes shaping genomic instability in cancer. The combined results point to G4 DNA, activation of cell cycle/DNA repair pathways, and mitochondrial dysfunction as three major factors driving the accumulation of somatic mutations in cancer cells.
Collapse
Affiliation(s)
- Albino Bacolla
- Departments of Cancer Biology and of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, 6767 Bertner Avenue, Houston, TX, 77030, USA.
| | - Zu Ye
- Departments of Cancer Biology and of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, 6767 Bertner Avenue, Houston, TX, 77030, USA.
| | - Zamal Ahmed
- Departments of Cancer Biology and of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, 6767 Bertner Avenue, Houston, TX, 77030, USA.
| | - John A Tainer
- Departments of Cancer Biology and of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, 6767 Bertner Avenue, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Dalla Pozza E, Dando I, Pacchiana R, Liboi E, Scupoli MT, Donadelli M, Palmieri M. Regulation of succinate dehydrogenase and role of succinate in cancer. Semin Cell Dev Biol 2019; 98:4-14. [PMID: 31039394 DOI: 10.1016/j.semcdb.2019.04.013] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 01/08/2023]
Abstract
Succinate dehydrogenase (SDH) has been classically considered a mitochondrial enzyme with the unique property to participate in both the citric acid cycle and the electron transport chain. However, in recent years, several studies have highlighted the role of the SDH substrate, i.e. succinate, in biological processes other than metabolism, tumorigenesis being the most remarkable. For this reason, SDH has now been defined a tumor suppressor and succinate an oncometabolite. In this review, we discuss recent findings regarding alterations in SDH activity leading to succinate accumulation, which include SDH mutations, regulation of mRNA expression, post-translational modifications and endogenous SDH inhibitors. Further, we report an extensive examination of the role of succinate in cancer development through the induction of epigenetic and metabolic alterations and the effects on epithelial to mesenchymal transition, cell migration and invasion, and angiogenesis. Finally, we have focused on succinate and SDH as diagnostic markers for cancers having altered SDH expression/activity.
Collapse
Affiliation(s)
- Elisa Dalla Pozza
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Elio Liboi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy; Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, Verona, Italy.
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| | - Marta Palmieri
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
28
|
The loss of succinate dehydrogenase B expression is frequently identified in hemangioblastoma of the central nervous system. Sci Rep 2019; 9:5873. [PMID: 30971719 PMCID: PMC6458311 DOI: 10.1038/s41598-019-42338-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 03/27/2019] [Indexed: 12/23/2022] Open
Abstract
Succinate dehydrogenase (SDH) is a mitochondrial enzyme that plays an important role in both the Krebs cycle and the electron transport chain. SDH inactivation is associated with tumorigenesis in certain types of tumor. SDH consists of subunits A, B, C and D (SDHA, SDHB, SDHC, and SDHD, respectively). Immunohistochemistry for SDHB is a reliable method for detecting the inactivation of SDH by mutations in SDHA, SDHB, SDHC, SDHD and SDH complex assembly factor 2 (SDHAF2) genes with high sensitivity and specificity. SDHB immunohistochemistry has been used to examine the inactivation of SDH in various types of tumors. However, data on central nervous system (CNS) tumors are very limited. In the present study, we investigated the loss of SDHB immunoexpression in 90 cases of CNS tumors. Among the 90 cases of CNS tumors, only three cases of hemangioblastoma showed loss of SDHB immunoexpression. We further investigated SDHB immunoexpression in 35 cases of hemangioblastoma and found that 28 (80%) showed either negative or weak-diffuse pattern of SDHB immunoexpression, which suggests the inactivation of SDH. Our results suggest that SDH inactivation may represent an alternative pathway in the tumorigenesis of hemangioblastoma.
Collapse
|
29
|
Fan F, Sam R, Ryan E, Alvarado K, Villa-Cuesta E. Rapamycin as a potential treatment for succinate dehydrogenase deficiency. Heliyon 2019; 5:e01217. [PMID: 30805566 PMCID: PMC6374580 DOI: 10.1016/j.heliyon.2019.e01217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 11/21/2022] Open
Abstract
Drosophila melanogaster is a powerful model to study mitochondrial respiratory chain defects, particularly succinate dehydrogenase (SDH) deficiency. Mutations in sdh genes cause degenerative disorders and often lead to death. Therapies for such pathologies are based on a combination of vitamins and dietary supplements, and are rarely effective. In Drosophila, mutations in several of the genes encoding SDH resemble the pathology of SDH deficiency in humans, enabling the Drosophila model to be used in finding treatments for this condition. Here we show that exposure to the drug rapamycin improves the survival of sdh mutant strains, the activity of SDH and the impaired climbing associated with sdh mutations. However, the production of reactive oxygen species, the oxygen consumption of isolated mitochondria and the resistance to hyperoxia were minimally affected. Our results contribute to the current research seeking a treatment for mitochondrial disease.
Collapse
Affiliation(s)
- Frances Fan
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | - Rheba Sam
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | - Emma Ryan
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | | | - Eugenia Villa-Cuesta
- Biology Department, Adelphi University, Garden City, NY, USA
- NYU Winthrop Research Institute, Mineola, NY, USA
| |
Collapse
|