1
|
Makgoo L, Mosebi S, Mbita Z. Susceptibility of HPV-18 Cancer Cells to HIV Protease Inhibitors. Viruses 2024; 16:1622. [PMID: 39459955 PMCID: PMC11512214 DOI: 10.3390/v16101622] [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: 09/20/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cervical cancer cases continue to rise despite all the advanced screening and preventative measures put in place, which include human papillomavirus (HPV) vaccination. These soaring numbers can be attributed to the lack of effective anticancer drugs against cervical cancer; thus, repurposing the human immunodeficiency virus protease inhibitors is an attractive innovation. Therefore, this work was aimed at evaluating the potential anticancer activities of HIV-PIs against cervical cancer cells. The MTT viability assay was used to evaluate the effect of HIV protease inhibitors on the viability of cervical cancer cells (HeLa) and non-cancerous cells (HEK-293). Further confirmation of the MTT assay was performed by confirming the IC50s of these HIV protease inhibitors on cervical cancer cells and non-cancerous cells using the Muse™ Count and Viability assay. To confirm the mode of death induced by HIV protease inhibitors in the HPV-associated cervical cancer cell line, apoptosis was performed using Annexin V assay. In addition, the Muse™ Cell Cycle assay was used to check whether the HIV protease inhibitors promote or halt cell cycle progression in cervical cancer cells. HIV protease inhibitors did not affect the viability of non-cancerous cells (HEK-293), but they decreased the viability of HeLa cervical cancer cells in a dose-dependent manner. HIV protease inhibitors induced apoptosis in HPV-related cervical cancer cells. Furthermore, they also induced cell cycle arrest, thus halting cell cycle progression. Therefore, the use of HIV drugs, particularly HIV-1 protease inhibitors, as potential cancer therapeutics represents a promising strategy. This is supported by our study demonstrating their anticancer properties, notably in HPV-associated cervical cancer cell line.
Collapse
Affiliation(s)
- Lilian Makgoo
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X 1106, Sovenga, Polokwane 0727, South Africa;
| | - Salerwe Mosebi
- Department of Life and Consumer Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa;
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X 1106, Sovenga, Polokwane 0727, South Africa;
| |
Collapse
|
2
|
Shaji UP, Tuti N, Alim SK, Mohan M, Das S, Meur G, Swamy MJ, Anindya R. Inhibition of human DNA alkylation damage repair enzyme ALKBH2 by HIV protease inhibitor ritonavir. DNA Repair (Amst) 2024; 141:103732. [PMID: 39094381 DOI: 10.1016/j.dnarep.2024.103732] [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: 01/10/2024] [Revised: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
The human DNA repair enzyme AlkB homologue-2 (ALKBH2) repairs methyl adducts from genomic DNA and is overexpressed in several cancers. However, there are no known inhibitors available for this crucial DNA repair enzyme. The aim of this study was to examine whether the first-generation HIV protease inhibitors having strong anti-cancer activity can be repurposed as inhibitors of ALKBH2. We selected four such inhibitors and performed in vitro binding analysis against ALKBH2 based on alterations of its intrinsic tryptophan fluorescence and differential scanning fluorimetry. The effect of these HIV protease inhibitors on the DNA repair activity of ALKBH2 was also evaluated. Interestingly, we observed that one of the inhibitors, ritonavir, could inhibit ALKBH2-mediated DNA repair significantly via competitive inhibition and sensitized cancer cells to alkylating agent methylmethane sulfonate (MMS). This work may provide new insights into the possibilities of utilizing HIV protease inhibitor ritonavir as a DNA repair antagonist.
Collapse
Affiliation(s)
- Unnikrishnan P Shaji
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Sanga Reddy, Telangana 502284, India
| | - Nikhil Tuti
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Sanga Reddy, Telangana 502284, India
| | - S K Alim
- School of Chemistry, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Monisha Mohan
- Department of Science and Humanities, Indian Institute of Information Technology Design and Manufacturing (IIIT-DM) Kancheepuram, Chennai, Tamil Nadu 600127, India
| | - Susmita Das
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Sanga Reddy, Telangana 502284, India
| | - Gargi Meur
- ICMR-National Institute of Nutrition, Hyderabad, Telangana 500007, India
| | - Musti J Swamy
- School of Chemistry, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Roy Anindya
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Sanga Reddy, Telangana 502284, India.
| |
Collapse
|
3
|
Sgadari C, Scoppio B, Picconi O, Tripiciano A, Gaiani FM, Francavilla V, Arancio A, Campagna M, Palladino C, Moretti S, Monini P, Brambilla L, Ensoli B. Clinical Efficacy of the HIV Protease Inhibitor Indinavir in Combination with Chemotherapy for Advanced Classic Kaposi Sarcoma Treatment: A Single-Arm, Phase II Trial in the Elderly. CANCER RESEARCH COMMUNICATIONS 2024; 4:2112-2122. [PMID: 39028943 PMCID: PMC11324028 DOI: 10.1158/2767-9764.crc-24-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/04/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Kaposi sarcoma is a rare angioproliferative disease associated with human herpes virus-8 (HHV-8) infection. Kaposi sarcoma is frequent and aggressive in HIV-infected people, whereas the classic form (CKS) generally has an indolent course. Notably, all conventional therapies against Kaposi sarcoma have only temporary efficacy. We have previously shown that indinavir, a HIV protease-inhibitor with direct antiangiogenic and antitumor activity, is safe and effective in patients with early CKS, whereas effects are less prominent in advanced disease, probably due to the larger tumor mass. Therefore, the clinical response to indinavir was assessed in patients with advanced CKS after debulking chemotherapy. This was a monocentric phase 2 trial in elderly with progressive/advanced CKS treated with debulking chemotherapy and indinavir combined, followed by a maintenance phase with indinavir alone. Secondary endpoints included safety and Kaposi sarcoma biomarker evaluation.All evaluable patients (22) responded to debulking therapy. Out of these, 16 entered the indinavir maintenance phase. The overall response rate at end of maintenance was 75% (estimated median response-duration 43 months). Moreover, most responders showed further clinical improvements (lesion number/nodularity) during maintenance and post-treatment follow-up. Notably, after relapse, progressors did not require systemic Kaposi sarcoma therapy and showed clinical improvements (including disease stabilization) remaining on study. Responders also showed immune status amelioration with a consistent B-cell increase and positive changes of other biomarkers, including anti-HHV-8 natural killer activity. In advanced CKS a strategy combining indinavir and chemotherapy is safe and associated with high and durable response rates and it could be rapidly adopted for the clinical management of these patients. SIGNIFICANCE This phase-2 trial showed that the HIV protease inhibitor indinavir may boost and extend the duration of the effects of chemotherapy in elderly with advanced progressive classic Kaposi sarcoma, without additional toxicity. Further, the amelioration of the immune status seen in responders suggests a better control of HHV-8 infection and tumor-cell killing. Thus, indinavir combined with chemotherapy may represent an important tool for the clinical management of classic Kaposi sarcoma in elderly patients.
Collapse
Affiliation(s)
- Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Biancamaria Scoppio
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | | | - Francesca Maria Gaiani
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | | | - Angela Arancio
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Massimo Campagna
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Clelia Palladino
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| | - Lucia Brambilla
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
4
|
Xiong JX, Li YT, Tan XY, Chen T, Liu BH, Fu L. Targeting PRSS23 with tipranavir induces gastric cancer stem cell apoptosis and inhibits growth of gastric cancer via the MKK3/p38 MAPK-IL24 pathway. Acta Pharmacol Sin 2024; 45:405-421. [PMID: 37814123 PMCID: PMC10789761 DOI: 10.1038/s41401-023-01165-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
Gastric cancer stem cells (GCSCs) contribute to the refractory features of gastric cancer (GC) and are responsible for metastasis, relapse, and drug resistance. The key factors drive GCSC function and affect the clinical outcome of GC patients remain poorly understood. PRSS23 is a novel serine protease that is significantly up-regulated in several types of cancers and cancer stem cells, and related to tumor progression and drug resistance. In this study, we investigated the role of PRSS23 in GCSCs as well as the mechanism by which PRSS23 regulated the GCSC functions. We demonstrated that PRSS23 was critical for sustaining GCSC survival. By screening a collection of human immunodeficiency virus (HIV) protease inhibitors (PIs), we identified tipranavir as a PRSS23-targeting drug, which effectively killed both GCSC and GC cell lines (its IC50 values were 4.7 and 6.4 μM in GCSC1 cells and GCSC2 cells, respectively). Administration of tipranavir (25 mg·kg-1·d-1, i.p., for 8 days) in GCSC-derived xenograft mice markedly inhibited the growth of subcutaneous GCSC tumors without apparent toxicity. In contrast, combined treatment with 5-FU plus cisplatin did not affect the tumor growth but causing significant weight loss. Furthermore, we revealed that tipranavir induced GCSC cell apoptosis by suppressing PRSS23 expression, releasing MKK3 from the PRSS23/MKK3 complex to activate p38 MAPK, and thereby activating the IL24-mediated Bax/Bak mitochondrial apoptotic pathway. In addition, tipranavir was found to kill other types of cancer cell lines and drug-resistant cell lines. Collectively, this study demonstrates that by targeting both GCSCs and GC cells, tipranavir is a promising anti-cancer drug, and the clinical development of tipranavir or other drugs specifically targeting the PRSS23/MKK3/p38MAPK-IL24 mitochondrial apoptotic pathway may offer an effective approach to combat gastric and other cancers.
Collapse
Affiliation(s)
- Ji-Xian Xiong
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| | - Yu-Ting Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Xiang-Yu Tan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Tie Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Bao-Hua Liu
- Department of Biochemistry, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
5
|
Omar A, Marques N, Crawford N. Cancer and HIV: The Molecular Mechanisms of the Deadly Duo. Cancers (Basel) 2024; 16:546. [PMID: 38339297 PMCID: PMC10854577 DOI: 10.3390/cancers16030546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
The immune deficiency associated with human immunodeficiency virus (HIV) infection causes a distinct increased risk of developing certain cancer types. Kaposi sarcoma (KS), invasive cervical cancer and non-Hodgkin's lymphoma (NHL) are the prominent malignancies that manifest as a result of opportunistic viral infections in patients with advanced HIV infection. Despite the implementation of antiretroviral therapy (ART), the prevalence of these acquired immunodeficiency syndrome (AIDS)-defining malignancies (ADMs) remains high in developing countries. In contrast, developed countries have experienced a steady decline in the occurrence of these cancer types. However, there has been an increased mortality rate attributed to non-ADMs. Here, we provide a review of the molecular mechanisms that are responsible for the development of ADMs and non-ADMs which occur in HIV-infected individuals. It is evident that ART alone is not sufficient to fully mitigate the potential for ADMs and non-ADMs in HIV-infected individuals. To enhance the diagnosis and treatment of both HIV and malignancies, a thorough comprehension of the mechanisms driving the development of such cancers is imperative.
Collapse
Affiliation(s)
- Aadilah Omar
- Division of Oncology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | | | | |
Collapse
|
6
|
Chakraborty S, Ramasubbu K, Banerjee M, Balaji MP, Vinayagam Y, V DR. A systematic review on the molecular and clinical association between Human Papillomavirus and Human Immunodeficiency Virus co-infection in Head, Neck and Oral squamous cell carcinoma. Rev Med Virol 2023; 33:e2462. [PMID: 37280764 DOI: 10.1002/rmv.2462] [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: 03/27/2023] [Revised: 04/27/2023] [Accepted: 05/23/2023] [Indexed: 06/08/2023]
Abstract
Head and neck cancer, one of the most commonly prevalent malignancies globally is a complex category of tumours that comprises cancers of the oral cavity, pharynx, and larynx. A specific subgroup of such cancers has been found with some unique chromosomal, therapeutic, and epidemiologic traits with the possibility of affecting via co-infection. About 25% of all head and neck cancers in the population are human papillomavirus infection (HPV)-associated, typically developing in the oropharynx, which comprises the tonsils. In the period of efficient combined antiviral treatment, HPV-positive oral cancers are also becoming a significant contributor to illness and fatality for Human Immunodeficiency Virus (HIV)-infected persons. Although the prevalence and historical background of oral HPV transmission are not thoroughly understood, it seems likely that oral HPV transmission is relatively frequent in HIV-infected people when compared to the overall population. Therefore, there is a need to understand the mechanisms leading to this co-infection, as there is very little research related to that. Hence, this study mainly focus on the therapeutical and biomedical analysis of HPV and HIV co-infection in the above-mentioned cancer, including oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Shreya Chakraborty
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Kanagavalli Ramasubbu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Manosi Banerjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Menaka Priya Balaji
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Yamini Vinayagam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Devi Rajeswari V
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
Alaei M, Nazari SE, Pourali G, Asadnia A, Moetamani-Ahmadi M, Fiuji H, Tanzadehpanah H, Asgharzadeh F, Babaei F, Khojasteh-Leylakoohi F, Saeed Gataa I, Ali Kiani M, Ferns GA, Lam AKY, Hassanian SM, Khazaei M, Giovannetti E, Avan A. Therapeutic Potential of Targeting the Cytochrome P450 Enzymes Using Lopinavir/Ritonavir in Colorectal Cancer: A Study in Monolayers, Spheroids and In Vivo Models. Cancers (Basel) 2023; 15:3939. [PMID: 37568755 PMCID: PMC10417395 DOI: 10.3390/cancers15153939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Cytochrome P450 (CYP450) enzyme has been shown to be expressed in colorectal cancer (CRC) and its dysregulation is linked to tumor progression and a poor prognosis. Here we investigated the therapeutic potential of targeting CYP450 using lopinavir/ritonavir in CRC. The integrative systems biology method and RNAseq were utilized to investigate the differential levels of genes associated with patients with colorectal cancer. The antiproliferative activity of lopinavir/ritonavir was evaluated in both monolayer and 3-dimensional (3D) models, followed by wound-healing assays. The effectiveness of targeting CYP450 was examined in a mouse model, followed by histopathological analysis, biochemical tests (MDA, SOD, thiol, and CAT), and RT-PCR. The data of dysregulation expressed genes (DEG) revealed 1268 upregulated and 1074 down-regulated genes in CRC. Among the top-score genes and dysregulated pathways, CYPs were detected and associated with poor prognosis of patients with CRC. Inhibition of CYP450 reduced cell proliferation via modulating survivin, Chop, CYP13a, and induction of cell death, as detected by AnnexinV/PI staining. This agent suppressed the migratory behaviors of cells by induction of E-cadherin. Moreover, lopinavir/ritonavir suppressed tumor growth and fibrosis, which correlated with a reduction in SOD/thiol levels and increased MDA levels. Our findings illustrated the therapeutic potential of targeting the CYP450 using lopinavir/ritonavir in colorectal cancer, supporting future investigations on this novel therapeutic approach for the treatment of CRC.
Collapse
Affiliation(s)
- Maryam Alaei
- Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (M.A.); (S.M.H.)
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Seyedeh Elnaz Nazari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - AliReza Asadnia
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Mehrdad Moetamani-Ahmadi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
| | - Hamid Fiuji
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Hamid Tanzadehpanah
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad 91779-49367, Iran
| | - Fereshteh Asgharzadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
| | - Fatemeh Babaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
| | - Fatemeh Khojasteh-Leylakoohi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | | | - Mohammad Ali Kiani
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Gordon A. Ferns
- Department of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK;
| | - Alfred King-yin Lam
- Pathology, School of Medicine and Dentistry, Gold Coast Campus, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (M.A.); (S.M.H.)
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (H.F.); (M.A.K.)
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), 1081 HV Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per La Scienza, 56124 Pisa, Italy
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran; (S.E.N.); (G.P.); (A.A.); (M.M.-A.); (H.T.); (F.A.); (F.B.); (F.K.-L.); (M.K.)
- College of Medicine, University of Warith Al-Anbiyaa, Karbala 56001, Iraq;
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
8
|
Abou-Salim MA, Shaaban MA, Abd El Hameid MK, Alanazi MM, Halaweish F, Elshaier YAMM. Utilizing Estra-1,3,5,16-Tetraene Scaffold: Design and Synthesis of Nitric Oxide Donors as Chemotherapeutic Resistance Combating Agents in Liver Cancer. Molecules 2023; 28:molecules28062754. [PMID: 36985726 PMCID: PMC10055446 DOI: 10.3390/molecules28062754] [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: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
A new series of nitric oxide-releasing estra-1,3,5,16-tetraene analogs (NO-∆-16-CIEAs) was designed and synthesized as dual inhibitors for EGFR and MRP2 based on our previous findings on estra-1,3,5-triene analog NO-CIEA 17 against both HepG2 and HepG2-R cell lines. Among the target compounds, 14a (R-isomer) and 14b (S-isomer) displayed potent anti-proliferative activity against both HepG2 and HepG2-R cell lines in comparison to the reference drug erlotinib. Remarkably, compound 14a resulted in a prominent reduction in EGFR phosphorylation at a concentration of 1.20 µM with slight activity on the phosphorylation of MEK1/2 and ERK1/2. It also inhibits MRP2 expression in a dose-dependent manner with 24% inhibition and arrested the cells in the S phase of the cell cycle. Interestingly, compound 14a (estratetraene core) exhibited a twofold increase in anti-proliferative activity against both HepG2 and HepG2-R in comparison with the lead estratriene analog, demonstrating the significance of the designed ∆-16 unsaturation. The results shed a light on compound 14a and support further investigations to combat multidrug resistance in chemotherapy of hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Mahrous A Abou-Salim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohamed A Shaaban
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mohammed K Abd El Hameid
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fathi Halaweish
- Department of Chemistry and Biochemistry, South Dakota State University, Box 2202, Brookings, SD 57007, USA
| | - Yaseen A M M Elshaier
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32958, Egypt
| |
Collapse
|
9
|
Caddeo C, Miglionico R, Rinaldi R, Nigro I, Lamorte D, Chiummiento L, Lupattelli P, Funicello M, D’Orsi R, Valenti D, Santoro V, Fadda AM, Bisaccia F, Vassallo A, Armentano MF. PEGylated Liposomes Loaded with Carbamate Inhibitor ANP0903 Trigger Apoptosis by Enhancing ER Stress in HepG2 Cancer Cells. Int J Mol Sci 2023; 24:ijms24054552. [PMID: 36901980 PMCID: PMC10002784 DOI: 10.3390/ijms24054552] [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: 02/01/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Liver cancer is one of the most common causes of cancer death worldwide. In recent years, substantial progress has been made in the development of systemic therapies, but there is still the need for new drugs and technologies that can increase the survival and quality of life of patients. The present investigation reports the development of a liposomal formulation of a carbamate molecule, reported as ANP0903, previously tested as an inhibitor of HIV-1 protease and now evaluated for its ability to induce cytotoxicity in hepatocellular carcinoma cell lines. PEGylated liposomes were prepared and characterized. Small, oligolamellar vesicles were produced, as demonstrated by light scattering results and TEM images. The physical stability of the vesicles in biological fluids was demonstrated in vitro, alongside the stability during storage. An enhanced cellular uptake was verified in HepG2 cells treated with liposomal ANP0903, resulting in a greater cytotoxicity. Several biological assays were performed to elucidate the molecular mechanisms explaining the proapoptotic effect of ANP0903. Our results allow us to hypothesize that the cytotoxic action in tumor cells is probably due to the inhibition of the proteasome, resulting in an increase in the amount of ubiquitinated proteins within the cells, which in turn triggers activation of autophagy and apoptosis processes, resulting in cell death. The proposed liposomal formulation represents a promising approach to deliver a novel antitumor agent to cancer cells and enhance its activity.
Collapse
Affiliation(s)
- Carla Caddeo
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Rocchina Miglionico
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Roberta Rinaldi
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Ilaria Nigro
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy
| | - Lucia Chiummiento
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Paolo Lupattelli
- Department of Chimica, Sapienza University of Roma, p.le Aldo Moro 5, 00185 Roma, Italy
| | - Maria Funicello
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Rosarita D’Orsi
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Donatella Valenti
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Valentina Santoro
- Department of Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy
| | - Anna Maria Fadda
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Faustino Bisaccia
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Antonio Vassallo
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff TNcKILLERS s.r.l., Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Correspondence: ; Tel.: +39-0971205624
| | | |
Collapse
|
10
|
Leming AB, Johnston AL, Krempl GA, Fowle EJ, Morton DJ, Henson CE. Pathologic complete response following low-dose radiation for advanced oral cavity cancer in a patient with human immunodeficiency virus. J Otolaryngol Head Neck Surg 2022; 51:37. [PMID: 36192808 PMCID: PMC9531370 DOI: 10.1186/s40463-022-00586-6] [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: 11/16/2021] [Accepted: 07/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Advanced squamous cell carcinoma (SCCa) of the oral cavity is often not amenable to curative-intent therapy due to tumor location, tumor size, or comorbidities. CASE PRESENTATION A 51-year-old male patient with human immunodeficiency virus and on highly active antiretroviral therapy (HAART) presented with a cT4aN2c SCCa of the tongue. He received a preoperative single course of Quad-Shot radiation therapy to 14 Gy in 4 fractions followed by surgical resection. Patient had no residual carcinoma on surgical pathology and no evidence of disease on subsequent clinical and radiological exams. CONCLUSIONS To our knowledge, this is the first case of pathologic complete response for a patient on HAART following a single cycle of the Quad-Shot regimen for advanced oral cavity SCCa. Protease inhibitors in HAART can induce spontaneous tumor regression via inhibition of proteasome function and activation of apoptosis, and thus act as a cancer therapeutic.
Collapse
Affiliation(s)
- Amy B. Leming
- grid.266902.90000 0001 2179 3618College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ,grid.267308.80000 0000 9206 2401Present Address: Department of Otorhinolaryngology – Head and Neck Surgery, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Andrea L. Johnston
- grid.266902.90000 0001 2179 3618Department of Radiation Oncology, University of Oklahoma Health Sciences Center, 800 NE 10th Street, L100, Oklahoma City, OK 73104 USA
| | - Greg A. Krempl
- grid.266902.90000 0001 2179 3618Department of Otorhinolaryngology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Evan J. Fowle
- grid.266902.90000 0001 2179 3618Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Daniel J. Morton
- grid.266902.90000 0001 2179 3618Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ,grid.266902.90000 0001 2179 3618Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Christina E. Henson
- grid.266902.90000 0001 2179 3618Department of Radiation Oncology, University of Oklahoma Health Sciences Center, 800 NE 10th Street, L100, Oklahoma City, OK 73104 USA ,grid.266902.90000 0001 2179 3618Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| |
Collapse
|
11
|
Makgoo L, Mosebi S, Mbita Z. Molecular Mechanisms of HIV Protease Inhibitors Against HPV-Associated Cervical Cancer: Restoration of TP53 Tumour Suppressor Activities. Front Mol Biosci 2022; 9:875208. [PMID: 35620479 PMCID: PMC9127998 DOI: 10.3389/fmolb.2022.875208] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is a Human Papilloma virus-related disease, which is on the rise in a number of countries, globally. Two essential oncogenes, E6 and E7, drive cell transformation and cancer development. These two oncoproteins target two of the most important tumour suppressors, p53 and pRB, for degradation through the ubiquitin ligase pathway, thus, blocking apoptosis activation and deregulation of cell cycle. This pathway can be exploited for anticancer therapeutic interventions, and Human Immunodeficiency Virus Protease Inhibitors (HIV-PIs) have attracted a lot of attention for this anticancer drug development. HIV-PIs have proven effective in treating HPV-positive cervical cancers and shown to restore impaired or deregulated p53 in HPV-associated cervical cancers by inhibiting the 26S proteasome. This review will evaluate the role players, such as HPV oncoproteins involved cervical cancer development and how they are targeted in HIV protease inhibitors-induced p53 restoration in cervical cancer. This review also covers the therapeutic potential of HIV protease inhibitors and molecular mechanisms behind the HIV protease inhibitors-induced p53-dependent anticancer activities against cervical cancer.
Collapse
Affiliation(s)
- Lilian Makgoo
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| | - Salerwe Mosebi
- Department of Life and Consumer Sciences, University of South Africa, Florida, South Africa
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
- *Correspondence: Zukile Mbita,
| |
Collapse
|
12
|
Pyruvate kinase L/R links metabolism dysfunction to neuroendocrine differentiation of prostate cancer by ZBTB10 deficiency. Cell Death Dis 2022; 13:252. [PMID: 35306527 PMCID: PMC8934352 DOI: 10.1038/s41419-022-04694-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 12/05/2022]
Abstract
Neuroendocrine differentiation (NED) frequently occurs in androgen-deprivation therapy (ADT)-resistant prostate cancer (PCa) and is typically associated with metabolic pathway alterations, acquisition of lineage plasticity, and malignancy. There is no conventional therapeutic approach for PCa patients with NED pathologic features because the molecular targets are unknown. Here, we evaluated the regulatory mechanism of NED-associated metabolic reprogramming induced by ADT. We detected that the loss of the androgen-responsive transcription factor, zinc finger, and BTB domain containing 10 (ZBTB10), can activate pyruvate kinase L/R (PKLR) to enhance a NED response that is associated with glucose uptake by PCa cells. PKLR exhibits a tumor-promoting effect in PCa after ADT, but ZBTB10 can compensate for the glucose metabolism and NED capacity of PKLR through the direct transcriptional downregulation of PKLR. Targeting PKLR by drug repurposing with FDA-approved compounds can reduce the aggressiveness and NED of ADT-resistant PCa. We demonstrated that PKLR acts as a modulator to activate NED in PCa enhancement by loss of ZBTB10, thereby enabling PCa cells to mount a glycolysis response essential for therapeutic resistance. Our findings highlight the broad relation between NED and metabolic dysfunction to provide gene expression-based biomarkers for NEPC treatment.
Collapse
|
13
|
Lee Y, Oh C, Kim J, Park MS, Bae WK, Yoo KH, Hong S. Bioinspired nonheme iron complex that triggers mitochondrial apoptotic signalling pathway specifically for colorectal cancer cells. Chem Sci 2022; 13:737-747. [PMID: 35173938 PMCID: PMC8768841 DOI: 10.1039/d1sc05094j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
The activation of dioxygen is the keystone of all forms of aerobic life. Many biological functions rely on the redox versatility of metal ions to perform reductive activation-mediated processes entailing dioxygen and its partially reduced species including superoxide, hydrogen peroxide, and hydroxyl radicals, also known as reactive oxygen species (ROS). In biomimetic chemistry, a number of synthetic approaches have sought to design, synthesize and characterize reactive intermediates such as the metal-superoxo, -peroxo, and -oxo species, which are commonly found as key intermediates in the enzymatic catalytic cycle. However, the use of these designed complexes and their corresponding intermediates as potential candidates for cancer therapeutics has scarcely been endeavored. In this context, a series of biomimetic first-row transition metal complexes bearing a picolylamine-based water-soluble ligand, [M(HN3O2)]2+ (M = Mn2+, Fe2+, Co2+, Cu2+; HN3O2 = 2-(2-(bis(pyridin-2-ylmethyl)amino)ethoxy)ethanol) were synthesized and characterized by various spectroscopic methods including X-ray crystallography and their dioxygen and ROS activation reactivity were evaluated in situ and in vitro. It turned out that among these metal complexes, the iron complex, [Fe(HN3O2)(H2O)]2+, was capable of activating dioxygen and hydrogen peroxide and produced the ROS species (e.g., hydroxyl radical). Upon the incubation of these complexes with different cancer cells, such as cervical, breast, and colorectal cancer cells (MDA-MB-231, AU565, SK-BR-3, HeLa S3, HT-29, and HCT116 cells), only the iron complex triggered cellular apoptosis specifically for colorectal cancer cells; the other metal complexes show negligible anti-proliferative activity. More importantly, the biomimetic complexes were harmless to normal cells and produced less ROS therein. The use of immunocytochemistry combined with western blot analysis strongly supported that apoptosis occurred via the intrinsic mitochondrial pathway; in the intracellular network, [Fe(HN3O2)(H2O)]2+ resulted in (i) the activation and/or production of ROS species, (ii) the induction of intracellular impaired redox balance, and (iii) the promotion of the mitochondrial apoptotic signaling pathway in colorectal cancer cells. The results have implications for developing novel biomimetic complexes in cancer treatments and for designing potent candidates with cancer-specific antitumor activity. A water-soluble iron complex that produces hydroxyl radical species triggers colorectal cancer cell death via the mitochondrial apoptotic pathway.![]()
Collapse
Affiliation(s)
- Yool Lee
- Department of Chemistry, Sookmyung Women's University Seoul 04310 Korea
| | - Chaeun Oh
- Department of Biological Sciences, Sookmyung Women's University Seoul 04310 Korea
| | - Jin Kim
- Department of Chemistry, Sunchon National University Suncheon 57922 Korea
| | - Myong-Suk Park
- Division of Hemato-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital Hwasun Republic of Korea
| | - Woo Kyun Bae
- Division of Hemato-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital Hwasun Republic of Korea .,Combinatorial Tumor Immunotherapy MRC Center, Chonnam National University Medical School Hwasun Republic of Korea
| | - Kyung Hyun Yoo
- Department of Biological Sciences, Sookmyung Women's University Seoul 04310 Korea
| | - Seungwoo Hong
- Department of Chemistry, Sookmyung Women's University Seoul 04310 Korea
| |
Collapse
|
14
|
Nelfinavir Induces Cytotoxicity towards High-Grade Serous Ovarian Cancer Cells, Involving Induction of the Unfolded Protein Response, Modulation of Protein Synthesis, DNA Damage, Lysosomal Impairment, and Potentiation of Toxicity Caused by Proteasome Inhibition. Cancers (Basel) 2021; 14:cancers14010099. [PMID: 35008264 PMCID: PMC8750028 DOI: 10.3390/cancers14010099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC) accounts for 70% of all ovarian-cancer-related deaths. Mainstay treatment with platinum-based drugs following surgery results in favorable outcomes in the majority of patients; however, in >80% of cases, the disease relapses with eventual drug resistance. As such, urgent development of improved alternative therapies is necessary for HGSOC patients with lower life expectancy. Rapid repurposing of market available drugs for cancer therapy is a cost-effective alternative to bypass the decade-long traditional drug development pipeline. Among potential drug-repurposing candidates, nelfinavir (NFV)—an anti-infective agent to treat acquired immunodeficiency syndrome (AIDS)—has shown anti-cancer effects against diverse cancers; however, its remedial benefits against HGSOC are unknown. In this study, we explored how NFV targets HGSOC cells obtained from patients at platinum-sensitive and -resistant stages. We observed beneficial efficacy elicited by NFV against HGSOC in both disease conditions through multiple mechanistic avenues, suggesting positive drug-repurposing prospects. Abstract High-grade serous ovarian cancer (HGSOC) is a significant cause of mortality among women worldwide. Traditional treatment consists of platinum-based therapy; however, rapid development of platinum resistance contributes to lower life expectancy, warranting newer therapies to supplement the current platinum-based protocol. Repurposing market-available drugs as cancer therapeutics is a cost- and time-effective way to avail new therapies to drug-resistant patients. The anti-HIV agent nelfinavir (NFV) has shown promising toxicity against various cancers; however, its role against HGSOC is unknown. Here, we studied the effect of NFV against HGSOC cells obtained from patients along disease progression and carrying different sensitivities to platinum. NFV triggered, independently of platinum sensitivity, a dose-dependent reduction in the HGSOC cell number and viability, and a parallel increase in hypo-diploid DNA content. Moreover, a dose-dependent reduction in clonogenic survival of cells escaping the acute toxicity was indicative of long-term residual damage. In addition, dose- and time-dependent phosphorylation of H2AX indicated NFV-mediated DNA damage, which was associated with decreased survival and proliferation signals driven by the AKT and ERK pathways. NFV also mediated a dose-dependent increase in endoplasmic reticulum stress-related molecules associated with long-term inhibition of protein synthesis and concurrent cell death; such events were accompanied by a proapoptotic environment, signaled by increased phospho-eIF2α, ATF4, and CHOP, increased Bax/Bcl-2 ratio, and cleaved executer caspase-7. Finally, we show that NFV potentiates the short-term cell cycle arrest and long-term toxicity caused by the proteasome inhibitor bortezomib. Overall, our in vitro study demonstrates that NFV can therapeutically target HGSOC cells of differential platinum sensitivities via several mechanisms, suggesting its prospective repurposing benefit considering its good safety profile.
Collapse
|
15
|
Maze EA, Agit B, Reeves S, Hilton DA, Parkinson DB, Laraba L, Ercolano E, Kurian KM, Hanemann CO, Belshaw RD, Ammoun S. Human endogenous retrovirus type K promotes proliferation and confers sensitivity to anti-retroviral drugs in Merlin-negative schwannoma and meningioma. Cancer Res 2021; 82:235-247. [PMID: 34853069 DOI: 10.1158/0008-5472.can-20-3857] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/04/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022]
Abstract
Deficiency of the tumour suppressor Merlin causes development of schwannoma, meningioma, and ependymoma tumours, which can occur spontaneously or in the hereditary disease neurofibromatosis type 2 (NF2). Merlin mutations are also relevant in a variety of other tumours. Surgery and radiotherapy are current first-line treatments; however, tumours frequently recur with limited treatment options. Here, we use human Merlin-negative schwannoma and meningioma primary cells to investigate the involvement of the endogenous retrovirus HERV-K in tumour development. HERV-K proteins previously implicated in tumorigenesis were overexpressed in schwannoma and all meningioma grades, and disease-associated CRL4DCAF1 and YAP/TEAD pathways were implicated in this overexpression. In normal Schwann cells, ectopic overexpression of HERV-K Env increased proliferation and upregulated expression of c-Jun and pERK1/2, which are key components of known tumorigenic pathways in schwannoma, JNK/c-Jun and RAS/RAF/MEK/ERK. Furthermore, FDA-approved retroviral protease inhibitors ritonavir, atazanavir, and lopinavir reduced proliferation of schwannoma and grade I meningioma cells. These results identify HERV-K as a critical regulator of progression in Merlin-deficient tumours and offer potential strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Emmanuel A Maze
- School of Biomedical Sciences, Faculty of Health Medicine, Dentistry and Human Sciences, Plymouth University
| | - Bora Agit
- Faculty of Health Medicine, Dentistry and Human Sciences, Plymouth University
| | - Shona Reeves
- Faculty of Health Medicine, Dentistry and Human Sciences, Plymouth University
| | - David A Hilton
- Faculty of Health Medicine, Dentistry and Human Sciences, Plymouth University
| | - David B Parkinson
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry
| | - Liyam Laraba
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry
| | | | - Kathreena M Kurian
- Department of Neuropathology, Brain Tumour Research Group, Frenchay Hospital, University of Bristol
| | - C Oliver Hanemann
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry
| | | | - Sylwia Ammoun
- Faculty of Health Medicine, Dentistry and Human Sciences, Plymouth University
| |
Collapse
|
16
|
Two Novel Precursors of the HIV-1 Protease Inhibitor Darunavir Target the UPR/Proteasome System in Human Hepatocellular Carcinoma Cell Line HepG2. Cells 2021; 10:cells10113052. [PMID: 34831275 PMCID: PMC8618555 DOI: 10.3390/cells10113052] [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: 09/09/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Several pre-clinical and clinical reports suggest that HIV-1 protease inhibitors, in addition to the antiretroviral properties, possess pleiotropic pharmacological effects including anticancer action. Therefore, we investigated the pro-apoptotic activity in tumor cells of two molecules, RDD-19 and RDD-142, which are hydroxyethylamine derivatives’ precursors of darunavir and several HIV-1 protease inhibitors. Methods: Three hepatoma cell lines and one non-pathological cell line were treated with RDD-19 and RDD-142, and cell viability was assessed. The expression levels of several markers for ER stress, autophagy, cellular ubiquitination, and Akt activation were quantified in HepG2 cells treated with RDD-19 and RDD-142 to evaluate apoptotic and non-apoptotic cell death. Results: RDD-19 and RDD-142 showed a greater dose-dependent cytotoxicity towards the hepatic tumor cell line HepG2 compared to the non-pathological hepatic cell line IHH. Both molecules caused two types of cell death, a caspase-dependent apoptosis, which was ascertained by a series of biochemical and morphological assays, and a caspase-independent death that was characterized by the induction of ER stress and autophagy. The strong increase of ubiquitinated proteins inside the cells suggested that the target of these molecules could be the proteasome and in silico molecular docking analysis that was used to support the plausibility of this hypothesis. Furthermore, cells treated with the two compounds displayed decreased levels of p-AKT, which interferes with cell survival and proliferation. Conclusions: These findings demonstrate that two compounds, RDD-19 and RDD-142, have pleiotropic effects and that they may represent promising anticancer candidates.
Collapse
|
17
|
Gul S, Ozcan O, Asar S, Okyar A, Barıs I, Kavakli IH. In silico identification of widely used and well-tolerated drugs as potential SARS-CoV-2 3C-like protease and viral RNA-dependent RNA polymerase inhibitors for direct use in clinical trials. J Biomol Struct Dyn 2021; 39:6772-6791. [PMID: 32752938 PMCID: PMC7484590 DOI: 10.1080/07391102.2020.1802346] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Despite strict measures taken by many countries, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be an issue of global concern. Currently, there are no clinically proven pharmacotherapies for coronavirus disease 2019, despite promising initial results obtained from drugs such as azithromycin and hydroxychloroquine. Therefore, the repurposing of clinically approved drugs for use against SARS-CoV-2 has become a viable strategy. Here, we searched for drugs that target SARS-CoV-2 3C-like protease (3CLpro) and viral RNA-dependent RNA polymerase (RdRp) by in silico screening of the U.S. Food and Drug Administration approved drug library. Well-tolerated and widely used drugs were selected for molecular dynamics (MD) simulations to evaluate drug-protein interactions and their persistence under physiological conditions. Tetracycline, dihydroergotamine, ergotamine, dutasteride, nelfinavir, and paliperidone formed stable interactions with 3CLpro based on MD simulation results. Similar analysis with RdRp showed that eltrombopag, tipranavir, ergotamine, and conivaptan bound to the enzyme with high binding free energies. Docking results suggest that ergotamine, dihydroergotamine, bromocriptine, dutasteride, conivaptan, paliperidone, and tipranavir can bind to both enzymes with high affinity. As these drugs are well tolerated, cost-effective, and widely used, our study suggests that they could potentially to be used in clinical trials for the treatment of SARS-CoV-2-infected patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Seref Gul
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Onur Ozcan
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Sinan Asar
- Department of Anesthesiology and Reanimation, Bakırköy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Alper Okyar
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Turkey
| | - Ibrahim Barıs
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Ibrahim Halil Kavakli
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| |
Collapse
|
18
|
Lopiccolo J, Kawabata S, Gills JJ, Dennis PA. Combining Nelfinavir With Chloroquine Inhibits In Vivo Growth of Human Lung Cancer Xenograft Tumors. In Vivo 2021; 35:141-145. [PMID: 33402459 DOI: 10.21873/invivo.12241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Nelfinavir is a human immunodeficiency virus protease inhibitor that is currently being repositioned as an anticancer drug. Chloroquine, an anti-malarial lysosomotropic drug, inhibits autophagy. It has been reported that the combination of nelfinavir and chloroquine significantly enhances endoplasmic reticulum (ER) stress and induces selective cell death in multiple cell line models (in vitro). MATERIALS AND METHODS We assessed the effects of the combination of these drugs on human NSCLC cell lines in vitro using cell proliferation assay and performed preclinical treatment studies using cell line-derived xenograft mouse models in vivo. RESULTS In vitro, this combination enhanced inhibition of NSCLC cell proliferation with increased proteotoxicity, including ER stress, and apoptosis. In vivo, the growth of human NSCLC xenograft tumors was inhibited, which correlated with increased apoptosis and induction of ER stress as well as NSCLC growth in vitro. CONCLUSION Our findings suggest that the induction of proteotoxicity provides a promising new target for developing anticancer drugs.
Collapse
Affiliation(s)
- Jaclyn Lopiccolo
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Shigeru Kawabata
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Joell J Gills
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Phillip A Dennis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| |
Collapse
|
19
|
Sharma S, Deep A, Sharma AK. Current Treatment for Cervical Cancer: An Update. Anticancer Agents Med Chem 2021; 20:1768-1779. [PMID: 32091347 DOI: 10.2174/1871520620666200224093301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/26/2019] [Accepted: 11/12/2019] [Indexed: 12/25/2022]
Abstract
Cervical cancer is the leading gynecologic health problem which is considered as the 4th most widespread tumour in women. The prevalence of this fatal ailment is emerging gradually across the globe as about 18.1 million new cancer cases have been reported in 2018. The predominance of cervical cancer has been significantly found in low and middle-income countries as cervical cancer ranks fourth for both incidence and mortality, conversely, there are no effective screening systems available. This mortal state is certainly influenced by exposure of human papillomavirus, dysregulation of caspase enzyme, elevated expression of Inhibitor Apoptotic Protein (IAP), overexpression of Vascular Endothelial Growth Factors (VEGF), active/passive smoking, and dysfunction of the immune system. Generally, the clinical trial on pipeline drugs leads to the development of some promising new therapies that are more effective than standard approaches and often unavailable outside of the clinical setting. Indeed, several biological interventions that can modulate the pathological cascade of cervical cancer are still under investigation. Thus, there is a need to further summarise the promising therapies for cervical cancer as we have accomplished in HER2-positive breast cancer by targeting HER2 therapies and immune checkpoint inhibitors in melanoma. The present report revealed the pharmacokinetic/ pharmacodynamics aspects of various pipeline drugs that are promising for the treatment of cervical cancer. Moreover, the study revealed the possible mechanism, adverse drug reaction, combined therapy and pleiotropic action of these under investigational drugs, which can further improve the therapeutic efficacy and restrict the imaginable harmful effects.
Collapse
Affiliation(s)
- Sombeer Sharma
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Aakash Deep
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Arun K Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Haryana-122413, India
| |
Collapse
|
20
|
Al-Bayati AI, Razzak Mahmood AA, Al-Mazaydeh ZA, Rammaha MS, Al-bayati RI, Alsoubani F, Tahtamouni LH. Synthesis, docking study, and in vitro anticancer evaluation of new flufenamic acid derivatives. PHARMACIA 2021. [DOI: 10.3897/pharmacia.68.e66788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Novel compounds (6–10) were synthesized and confirmed by spectroscopic analysis, including AT-IR, 1HNMR and CHNS. Their cytotoxic effect was evaluated by MTT assay against two cancer cell lines and two normal cell types. Compound 7 exhibited anticancer activity against MCF-7 breast cancer cell line (GI50 = 63.9 µg/ml, 148 µM), without any effect against A549 lung cancer cells, or the normal cells. Compound 7 caused cytotoxicity in MCF-7 breast cancer cells by apoptotic cell death, as suggested by fragmented nuclei after DAPI staining and agarose gel electrophoresis. In addition, treating MCF-7 cells with compound 7 resulted in an increase in the level of caspase 9 mRNA level, and its activation. Moreover, compound 7-treated MCF-7 cells showed enhanced cytochrome c release from the mitochondria to the cytosol, signifying an induction of the intrinsic apoptotic pathway. Finally, compound 7 exhibited epidermal growth factor receptor (EGFR) kinase inhibitory activity at (EC50 = 0.13 µM), which was matched by molecular docking studies that showed compound 7 might be an important EGFR kinase inhibitor.
Collapse
|
21
|
Marima R, Hull R, Dlamini Z, Penny C. The profiling, identification, quantification and analysis of differentially expressed genes (DEGs) in response to drug treatment in lung cancer. MethodsX 2021; 8:101381. [PMID: 34430277 PMCID: PMC8374453 DOI: 10.1016/j.mex.2021.101381] [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: 10/15/2020] [Accepted: 05/02/2021] [Indexed: 11/17/2022] Open
Abstract
The profiling and identification of genes that are differentially expressed is frequently used to underpin the underlying molecular mechanisms of biological conditions and provides a molecular foothold on biological questions of interest. However, this can be a daunting task since there is a cross talk and overlap of some of the components of the signalling pathways. The deregulation of the cell cycle signalling pathway is a hallmark of cancer, including lung cancer. Proper regulation of the cell cycle results in cellular homeostasis between cell proliferation and cell death. The comprehension of the cell cycle regulation in drug metabolism studies is of significance. This study aimed at elucidating the regulation of cell cycle genes' in response to LPV/r in lung cells. Thus, this study describes methodology for revealing molecular mechanisms employed by LPV/r to induce stress on genomic DNA. This approach is based on the interrogation of a panel of 84 genes related to the cell cycle pathway, and how the differentially expressed genes' expression pattern corroborates loss in nuclear integrity (phenotypic observation). MAD2L2, AURKB and CASP3 gene expressions were further confirmed by RT-qPCR. Furthermore, the use of in-silico bioinformatics tools integrates the molecular profiles and phenotypic changes. This approach revealed the activation of the DNA damage response (DDR) pathway in response to LPV/r treatment. The proposed methodology will aid in the comprehension of drug metabolism at genotypic and phenotypic levels.•Gene profiling often reveals the underlying molecular mechanisms.•RT2 PCR gene arrays have integrated patented quality controls and allow reliable gene expression analysis.•In-silico bioinformatics analysis help reveal pathways affected, that often correspond to phenotypic changes/features.
Collapse
Affiliation(s)
- Rahaba Marima
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV‐Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield 0028, South Africa
- Department of Internal Medicine, School of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| | - Rodney Hull
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV‐Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield 0028, South Africa
| | - Zodwa Dlamini
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV‐Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield 0028, South Africa
| | - Clement Penny
- Department of Internal Medicine, School of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| |
Collapse
|
22
|
Li S, Zhang F, Xiao X, Guo Y, Wen Z, Li M, Pu X. Prediction of Synergistic Drug Combinations for Prostate Cancer by Transcriptomic and Network Characteristics. Front Pharmacol 2021; 12:634097. [PMID: 33986671 PMCID: PMC8112211 DOI: 10.3389/fphar.2021.634097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/04/2021] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer (PRAD) is a major cause of cancer-related deaths. Current monotherapies show limited efficacy due to often rapidly emerging resistance. Combination therapies could provide an alternative solution to address this problem with enhanced therapeutic effect, reduced cytotoxicity, and delayed the appearance of drug resistance. However, it is prohibitively cost and labor-intensive for the experimental approaches to pick out synergistic combinations from the millions of possibilities. Thus, it is highly desired to explore other efficient strategies to assist experimental researches. Inspired by the challenge, we construct the transcriptomics-based and network-based prediction models to quickly screen the potential drug combination for Prostate cancer, and further assess their performance by in vitro assays. The transcriptomics-based method screens nine possible combinations. However, the network-based method gives discrepancies for at least three drug pairs. Further experimental results indicate the dose-dependent effects of the three docetaxel-containing combinations, and confirm the synergistic effects of the other six combinations predicted by the transcriptomics-based model. For the network-based predictions, in vitro tests give opposite results to the two combinations (i.e. mitoxantrone-cyproheptadine and cabazitaxel-cyproheptadine). Namely, the transcriptomics-based method outperforms the network-based one for the specific disease like Prostate cancer, which provide guideline for selection of the computational methods in the drug combination screening. More importantly, six combinations (the three mitoxantrone-containing and the three cabazitaxel-containing combinations) are found to be promising candidates to synergistically conquer Prostate cancer.
Collapse
Affiliation(s)
- Shiqi Li
- College of Chemistry, Sichuan University, Chengdu, China
| | - Fuhui Zhang
- College of Chemistry, Sichuan University, Chengdu, China
| | - Xiuchan Xiao
- School of Material Science and Environmental Engineering, Chengdu Technological University, Chengdu, China
| | - Yanzhi Guo
- College of Chemistry, Sichuan University, Chengdu, China
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, China
| | - Xuemei Pu
- College of Chemistry, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Li M, Liu Y, Deng Y, Pan L, Fu H, Han X, Li Y, Shi H, Wang T. Therapeutic potential of endogenous hydrogen sulfide inhibition in breast cancer (Review). Oncol Rep 2021; 45:68. [PMID: 33760221 PMCID: PMC8020202 DOI: 10.3892/or.2021.8019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Hydrogen sulfide (H2S), the third gas signal molecule, is associated with the modulation of various physiological and pathological processes. Recent studies have reevealed that endogenous H2S may promote proliferation, induce angiogenesis and inhibit apoptosis, thereby stimulating oncogenesis. Conversely, decreased endogenous H2S release suppresses growth of various tumors including breast cancer. This observation suggests an alternative tumor therapy strategy by inhibiting H2S-producing enzymes to reduce the release of endogenous H2S. Breast cancer is the most common type of cancer in women. Due to the lack of approved targeted therapy, its recurrence and metastasis still affect its clinical treatment. In recent years, significant progress has been made in the control of breast cancer by using inhibitors on H2S-producing enzymes. This review summarized the roles of endogenous H2S-producing enzymes in breast cancer and the effects of the enzyme inhibitors on anticancer and anti-metastasis, with the aim of providing new insights for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ming Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ya Liu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuying Deng
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Limin Pan
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Han Fu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Xue Han
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuxi Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Haimei Shi
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Tianxiao Wang
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
24
|
Haq H, Elyanu P, Bulsara S, Bacha JM, Campbell LR, El-Mallawany NK, Keating EM, Kisitu GP, Mehta PS, Rees CA, Slone JS, Kekitiinwa AR, Matshaba M, Mizwa MB, Mwita L, Schutze GE, Wanless SR, Scheurer ME, Lubega J. Association between Antiretroviral Therapy and Cancers among Children Living with HIV in Sub-Saharan Africa. Cancers (Basel) 2021; 13:cancers13061379. [PMID: 33803641 PMCID: PMC8003101 DOI: 10.3390/cancers13061379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/02/2021] [Accepted: 03/14/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Most children infected with HIV live in Sub-Sahara Africa (SSA). These children are at risk of cancers related to HIV infection, but the degree of this risk and how it is influenced by antiretroviral therapy (ART) is unknown. In this study, we determined the subtypes, incidence, and risk factors of cancers in children with HIV in SSA and receiving ART with the goal of learning how we may prevent these cancers. We found that Kaposi sarcoma and lymphoma are the most common, comprising about 77% and 19% of cancers in these children, respectively. For every 100,000 person-years, 47.6 children developed cancer. Waiting to start ART until after 2 years old and having had severe immunosuppression were the two biggest risk factors for cancer that we identified. The findings justify the recommendations to start children on ART as soon as they are diagnosed with HIV regardless of their CD4 immune status. Abstract Approximately 91% of the world’s children living with HIV (CLWH) are in sub-Saharan Africa (SSA). Living with HIV confers a risk of developing HIV-associated cancers. To determine the incidence and risk factors for cancer among CLWH, we conducted a nested case-control study of children 0–18 years from 2004–2014 at five centers in four SSA countries. Incident cases of cancer and HIV were frequency-matched to controls with HIV and no cancer. We calculated the incidence density by cancer type, logistic regression, and relative risk to evaluate risk factors of cancer. The adjusted incidence density of all cancers, Kaposi sarcoma, and lymphoma were 47.6, 36.6, and 8.94 per 100,000 person-years, respectively. Delayed ART until after 2 years of age was associated with cancer (OR = 2.71, 95% CI 1.51, 4.89) even after adjusting for World Health Organization clinical stage at the time of enrolment for HIV care (OR = 2.85, 95% CI 1.57, 5.13). The relative risk of cancer associated with severe CD4 suppression was 6.19 (p = 0.0002), 2.33 (p = 0.0042), and 1.77 (p = 0.0305) at 1, 5, and 10 years of ART, respectively. The study demonstrates the high risk of cancers in CLWH and the potential benefit of reducing this risk by the early initiation of ART.
Collapse
Affiliation(s)
- Heather Haq
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Peter Elyanu
- Baylor College of Medicine Children’s Foundation-Uganda, Kampala, Uganda; (P.E.); (G.P.K.); (A.R.K.)
| | - Shaun Bulsara
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
| | - Jason M. Bacha
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
- Baylor College of Medicine Children’s Foundation-Tanzania, Mbeya, Tanzania;
| | - Liane R. Campbell
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
- Baylor College of Medicine Children’s Foundation-Tanzania, Mbeya, Tanzania;
| | - Nader K. El-Mallawany
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
| | - Elizabeth M. Keating
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine Children’s Foundation-Lesotho, Maseru, Lesotho
| | - Grace P. Kisitu
- Baylor College of Medicine Children’s Foundation-Uganda, Kampala, Uganda; (P.E.); (G.P.K.); (A.R.K.)
| | - Parth S. Mehta
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Global Hematology Oncology Pediatric Excellence Program, Texas Children’s Cancer and Hematology Centers, Houston, TX 77030, USA
| | - Chris A. Rees
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine Children’s Foundation-Malawi, Lilongwe, Malawi
| | - Jeremy S. Slone
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Global Hematology Oncology Pediatric Excellence Program, Texas Children’s Cancer and Hematology Centers, Houston, TX 77030, USA
| | - Adeodata R. Kekitiinwa
- Baylor College of Medicine Children’s Foundation-Uganda, Kampala, Uganda; (P.E.); (G.P.K.); (A.R.K.)
| | - Mogomotsi Matshaba
- Botswana-Baylor Children’s Clinical Centre of Excellence, Gabarone, Botswana;
| | - Michael B. Mizwa
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Lumumba Mwita
- Baylor College of Medicine Children’s Foundation-Tanzania, Mbeya, Tanzania;
| | - Gordon E. Schutze
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Sebastian R. Wanless
- Baylor College of Medicine International Pediatric AIDS Initiative (BIPAI) at Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Michael E. Scheurer
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
| | - Joseph Lubega
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (H.H.); (S.B.); (J.M.B.); (L.R.C.); (N.K.E.-M.); (E.M.K.); (P.S.M.); (C.A.R.); (J.S.S.); (M.B.M.); (G.E.S.); (M.E.S.)
- Global Hematology Oncology Pediatric Excellence Program, Texas Children’s Cancer and Hematology Centers, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-8328224242
| |
Collapse
|
25
|
Park S, Auyeung A, Lee DL, Lambert PF, Carchman EH, Sherer NM. HIV-1 Protease Inhibitors Slow HPV16-Driven Cell Proliferation through Targeted Depletion of Viral E6 and E7 Oncoproteins. Cancers (Basel) 2021; 13:949. [PMID: 33668328 PMCID: PMC7956332 DOI: 10.3390/cancers13050949] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/09/2021] [Accepted: 02/20/2021] [Indexed: 02/05/2023] Open
Abstract
High-risk human papillomavirus strain 16 (HPV16) causes oral and anogenital cancers through the activities of two viral oncoproteins, E6 and E7, that dysregulate the host p53 and pRb tumor suppressor pathways, respectively. The maintenance of HPV16-positive cancers requires constitutive expression of E6 and E7. Therefore, inactivating these proteins could provide the basis for an anticancer therapy. Herein we demonstrate that a subset of aspartyl protease inhibitor drugs currently used to treat HIV/AIDS cause marked reductions in HPV16 E6 and E7 protein levels using two independent cell culture models: HPV16-transformed CaSki cervical cancer cells and NIKS16 organotypic raft cultures (a 3-D HPV16-positive model of epithelial pre-cancer). Treatment of CaSki cells with some (lopinavir, ritonavir, nelfinavir, and saquinavir) but not other (indinavir and atazanavir) protease inhibitors reduced E6 and E7 protein levels, correlating with increased p53 protein levels and decreased cell viability. Long-term (>7 day) treatment of HPV16-positive NIKS16 raft cultures with saquinavir caused epithelial atrophy with no discernible effects on HPV-negative rafts, demonstrating selectivity. Saquinavir also reduced HPV16's effects on markers of the cellular autophagy pathway in NIKS16 rafts, a hallmark of HPV-driven pre-cancers. Taken together, these data suggest HIV-1 protease inhibitors be studied further in the context of treating or preventing HPV16-positive cancers.
Collapse
Affiliation(s)
- Soyeong Park
- McArdle Laboratory for Cancer Research, Deptartment of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.P.); (D.L.L.); (P.F.L.)
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
| | - Andrew Auyeung
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Denis L. Lee
- McArdle Laboratory for Cancer Research, Deptartment of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.P.); (D.L.L.); (P.F.L.)
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, Deptartment of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.P.); (D.L.L.); (P.F.L.)
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
| | - Evie H. Carchman
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Nathan M. Sherer
- McArdle Laboratory for Cancer Research, Deptartment of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.P.); (D.L.L.); (P.F.L.)
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (A.A.); (E.H.C.)
| |
Collapse
|
26
|
Rahman MR, Islam T, Nicoletti F, Petralia MC, Ciurleo R, Fisicaro F, Pennisi M, Bramanti A, Demirtas TY, Gov E, Islam MR, Mussa BM, Moni MA, Fagone P. Identification of Common Pathogenetic Processes between Schizophrenia and Diabetes Mellitus by Systems Biology Analysis. Genes (Basel) 2021; 12:genes12020237. [PMID: 33562405 PMCID: PMC7916024 DOI: 10.3390/genes12020237] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder characterized by both positive symptoms (i.e., psychosis) and negative symptoms (such as apathy, anhedonia, and poverty of speech). Epidemiological data show a high likelihood of early onset of type 2 diabetes mellitus (T2DM) in SCZ patients. However, the molecular processes that could explain the epidemiological association between SCZ and T2DM have not yet been characterized. Therefore, in the present study, we aimed to identify underlying common molecular pathogenetic processes and pathways between SCZ and T2DM. To this aim, we analyzed peripheral blood mononuclear cell (PBMC) transcriptomic data from SCZ and T2DM patients, and we detected 28 differentially expressed genes (DEGs) commonly modulated between SCZ and T2DM. Inflammatory-associated processes and membrane trafficking pathways as common biological processes were found to be in common between SCZ and T2DM. Analysis of the putative transcription factors involved in the regulation of the DEGs revealed that STAT1 (Signal Transducer and Activator of Transcription 1), RELA (v-rel reticuloendotheliosis viral oncogene homolog A (avian)), NFKB1 (Nuclear Factor Kappa B Subunit 1), and ERG (ETS-related gene) are involved in the expression of common DEGs in SCZ and T2DM. In conclusion, we provide core molecular signatures and pathways that are shared between SCZ and T2DM, which may contribute to the epidemiological association between them.
Collapse
Affiliation(s)
- Md Rezanur Rahman
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh;
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Enayetpur, Sirajganj 6751, Bangladesh;
| | - Tania Islam
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Enayetpur, Sirajganj 6751, Bangladesh;
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.F.); (M.P.); (P.F.)
- Correspondence:
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.F.); (M.P.); (P.F.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.F.); (M.P.); (P.F.)
| | - Alessia Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Talip Yasir Demirtas
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Turkes Science and Technology University, Adana 01250, Turkey; (T.Y.D.); (E.G.)
| | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Turkes Science and Technology University, Adana 01250, Turkey; (T.Y.D.); (E.G.)
| | - Md Rafiqul Islam
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia;
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Bashair M. Mussa
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
| | - Mohammad Ali Moni
- WHO Collaborating Centre on eHealth, UNSW Digital Health, School of Public Health and Community Medicine, Faculty of Medicine, Sydney, NSW 2052, Australia;
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.F.); (M.P.); (P.F.)
| |
Collapse
|
27
|
Cabrera-Andrade A, López-Cortés A, Jaramillo-Koupermann G, González-Díaz H, Pazos A, Munteanu CR, Pérez-Castillo Y, Tejera E. A Multi-Objective Approach for Anti-Osteosarcoma Cancer Agents Discovery through Drug Repurposing. Pharmaceuticals (Basel) 2020; 13:ph13110409. [PMID: 33266378 PMCID: PMC7700154 DOI: 10.3390/ph13110409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor. Although nowadays 5-year survival rates can reach up to 60–70%, acute complications and late effects of osteosarcoma therapy are two of the limiting factors in treatments. We developed a multi-objective algorithm for the repurposing of new anti-osteosarcoma drugs, based on the modeling of molecules with described activity for HOS, MG63, SAOS2, and U2OS cell lines in the ChEMBL database. Several predictive models were obtained for each cell line and those with accuracy greater than 0.8 were integrated into a desirability function for the final multi-objective model. An exhaustive exploration of model combinations was carried out to obtain the best multi-objective model in virtual screening. For the top 1% of the screened list, the final model showed a BEDROC = 0.562, EF = 27.6, and AUC = 0.653. The repositioning was performed on 2218 molecules described in DrugBank. Within the top-ranked drugs, we found: temsirolimus, paclitaxel, sirolimus, everolimus, and cabazitaxel, which are antineoplastic drugs described in clinical trials for cancer in general. Interestingly, we found several broad-spectrum antibiotics and antiretroviral agents. This powerful model predicts several drugs that should be studied in depth to find new chemotherapy regimens and to propose new strategies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Correspondence: (A.C.-A.); (E.T.)
| | - Andrés López-Cortés
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), 28029 Madrid, Spain
| | - Gabriela Jaramillo-Koupermann
- Laboratorio de Biología Molecular, Subproceso de Anatomía Patológica, Hospital de Especialidades Eugenio Espejo, Quito 170403, Ecuador;
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, and Basque Center for Biophysics CSIC-UPV/EHU, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Alejandro Pazos
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Cristian R. Munteanu
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito 170125, Ecuador
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de Las Américas, Quito 170125, Ecuador
- Correspondence: (A.C.-A.); (E.T.)
| |
Collapse
|
28
|
The Anti-Cancer Properties of the HIV Protease Inhibitor Nelfinavir. Cancers (Basel) 2020; 12:cancers12113437. [PMID: 33228205 PMCID: PMC7699465 DOI: 10.3390/cancers12113437] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary To this day, cancer remains a medical challenge despite the development of cutting-edge diagnostic methods and therapeutics. Thus, there is a continual demand for improved therapeutic options for managing cancer patients. However, novel drug development requires decade-long time commitment and financial investments. Repurposing approved and market-available drugs for cancer therapy is a way to reduce cost and the timeframe for developing new therapies. Nelfinavir is an anti-infective agent that has extensively been used to treat acquired immunodeficiency syndrome (AIDS) in adult and pediatric patients. In addition to its anti-infective properties, nelfinavir has demonstrated potent off-target anti-cancer effects, suggesting that it could be a suitable candidate for drug repurposing for cancer. In this review, we systematically compiled the therapeutic benefits of nelfinavir against cancer as a single drug or in combination with chemoradiotherapy, and outlined the possible underlying mechanistic pathways contributing to the anti-cancer effects. Abstract Traditional cancer treatments may lose efficacy following the emergence of novel mutations or the development of chemoradiotherapy resistance. Late diagnosis, high-cost of treatment, and the requirement of highly efficient infrastructure to dispense cancer therapies hinder the availability of adequate treatment in low-income and resource-limited settings. Repositioning approved drugs as cancer therapeutics may reduce the cost and timeline for novel drug development and expedite the availability of newer, efficacious options for patients in need. Nelfinavir is a human immunodeficiency virus (HIV) protease inhibitor that has been approved and is extensively used as an anti-infective agent to treat acquired immunodeficiency syndrome (AIDS). Yet nelfinavir has also shown anti-cancer effects in in vitro and in vivo studies. The anti-cancer mechanism of nelfinavir includes modulation of different cellular conditions, such as unfolded protein response, cell cycle, apoptosis, autophagy, the proteasome pathway, oxidative stress, the tumor microenvironment, and multidrug efflux pumps. Multiple clinical trials indicated tolerable and reversible toxicities during nelfinavir treatment in cancer patients, either as a monotherapy or in combination with chemo- or radiotherapy. Since orally available nelfinavir has been a safe drug of choice for both adult and pediatric HIV-infected patients for over two decades, exploiting its anti-cancer off-target effects will enable fast-tracking this newer option into the existing repertoire of cancer chemotherapeutics.
Collapse
|
29
|
Marcia M, Vania B, Pruccoli G, Vallero SG, Barisone E, Scolfaro C, Fagioli F. Acute lymphoblastic leukemia onset in a 3-year-old child with COVID-19. Pediatr Blood Cancer 2020; 67:e28423. [PMID: 32706512 PMCID: PMC7404518 DOI: 10.1002/pbc.28423] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Marta Marcia
- Department of Pediatric and Public Health SciencesRegina Margherita Children's HospitalUniversity of TurinTurinItaly
| | - Barbara Vania
- Department of Pediatric and Public Health SciencesRegina Margherita Children's HospitalUniversity of TurinTurinItaly
| | - Giulia Pruccoli
- Department of Pediatric and Public Health SciencesRegina Margherita Children's HospitalUniversity of TurinTurinItaly
| | - Stefano G. Vallero
- Department of Pediatric Onco‐HematologyRegina Margherita Children's HospitalTurinItaly
| | - Elena Barisone
- Department of Pediatric Onco‐HematologyRegina Margherita Children's HospitalTurinItaly
| | - Carlo Scolfaro
- Department of Pediatric and Public Health SciencesInfectious Diseases UnitRegina Margherita Children's HospitalTurinItaly
| | - Franca Fagioli
- Department of Pediatric Onco‐HematologyRegina Margherita Children's HospitalTurinItaly
| |
Collapse
|
30
|
Marima R, Hull R, Dlamini Z, Penny C. The dual protease inhibitor lopinavir/ritonavir (LPV/r) exerts genotoxic stress on lung cells. Biomed Pharmacother 2020; 132:110829. [PMID: 33059259 DOI: 10.1016/j.biopha.2020.110829] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/06/2020] [Accepted: 09/28/2020] [Indexed: 01/13/2023] Open
Abstract
The Sub-Saharan countries, particularly South Africa has the largest number of people living with HIV, accompanied by the largest antiretroviral treatment (ART) programme in the world. The Highly Active Antiretroviral Treatment (HAART) is the most effective regimen against HIV/AIDS and has improved the lifespan and quality of life of HIV positive patients. HAART has also led to a decrease in the incidence of AIDS defining cancers (ADCs) while there is an increased incidence of the non-AIDS Defining Cancers (NADCs), such as lung cancer in the HAART era. The association between lung tumourigenesis and the use of HAART components such as the dual protease inhibitor (PI) lopinavir/ritonavir (LPV/r) is poorly understood. Using cell and molecular biological approaches, this study aimed at elucidating the effects of LPV/r on the regulation of the cell cycle related genes in normal (MRC-5) and adenocarcinoma (A549) lung cells. Initially, the nuclear integrity of these cells in response to LPV/r was determined using DAPI staining. The effect of LPV/r on cell cycle genes was evaluated through the use of a RT2 PCR gene array of 84 genes related to the cell cycle signaling pathway. The PCR array data was validated by Real-Time Quantification PCR (RT-qPCR). Ingenuity Pathway Analysis (IPA) bio-informatics tool was employed to disclose the molecular mechanism/s observed at cellular and gene expression levels. Loss of nuclear integrity and the upregulation of the p53 DNA damage response (DDR) pathway was revealed by DAPI staining, differential gene expression and IPA core analysis. Furthermore, MAD2L2 and AURKB which also play a role in the DDR pathway were shown to be differentially expressed. The activation of the CASP3 gene in response to LPV/r in A549 cells was also observed. The findings of this study suggest genotoxic properties of LPV/r in healthy normal lung fibroblasts cells and anti-tumour properties in the A549 cells.
Collapse
Affiliation(s)
- Rahaba Marima
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa; Department of Internal Medicine, School of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa.
| | - Rodney Hull
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa
| | - Zodwa Dlamini
- SAMRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa
| | - Clement Penny
- Department of Internal Medicine, School of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| |
Collapse
|
31
|
Marima R, Hull R, Dlamini Z, Penny C. Efavirenz and Lopinavir/Ritonavir Alter Cell Cycle Regulation in Lung Cancer. Front Oncol 2020; 10:1693. [PMID: 32984047 PMCID: PMC7484481 DOI: 10.3389/fonc.2020.01693] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022] Open
Abstract
Highly active anti-retroviral treatment (HAART) is currently the most effective treatment for HIV/AIDS. Additionally, HIV positive patients receiving HAART have a better health-related quality of life (HRQoL). Cancers previously associated with HIV/AIDS also known as the AIDS defining cancers (ADCs), such as Kaposi's sarcoma and non-Hodgkin's lymphoma have been on the decline since the introduction of HAART. However, non-AIDS defining cancers (NADCs), in particular, lung cancers have been documented to be on the rise. The association between the use of HAART components and lung carcinogenesis is poorly understood. This study aimed at elucidating the effects of two HAART components [efavirenz (EFV), and lopinavir/ritonavir (LPV/r)] on lung cancer. This was achieved through the use of in vitro cell biological approaches to assess cell health, including cell viability, Real Time Cell Analysis (RTCA) growth monitoring, evaluation of the cell cycle, and progression to apoptosis, following on drug treatments. At plasma level concentrations, both EFV and LPV/r induced S-phase arrest, while at lower concentrations both drugs promoted the progression of cells into G2/M phase following cell cycle FACS analysis. At higher concentrations although cell viability assays reflected anti-proliferative effects of the drugs, this was not statistically significant. RTCA showed a significant decline in cell viability in response to the highest dose of LPV/r. Dual staining by Annexin V-FITC and PI confirmed significant pro-apoptotic effects were promoted by LPV/r. Both EFV and LPV/r exert double-edged oncogenic effects on MRC-5 and A549 lung cells, acting to either promote cell proliferation or to enhance apoptosis. This is affected by EFV and LPV/r altering cell cycle progression, with a significant S-phase arrest, this being an indication of cellular stress, cytotoxicity, and DNA damage within the cell.
Collapse
Affiliation(s)
- Rahaba Marima
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| | - Rodney Hull
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa
| | - Zodwa Dlamini
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| |
Collapse
|
32
|
Hazafa A, ur-Rahman K, Haq IU, Jahan N, Mumtaz M, Farman M, Naeem H, Abbas F, Naeem M, Sadiqa S, Bano S. The broad-spectrum antiviral recommendations for drug discovery against COVID-19. Drug Metab Rev 2020; 52:408-424. [PMID: 32546018 PMCID: PMC7309307 DOI: 10.1080/03602532.2020.1770782] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Despite to outbreaks of highly pathogenic beta and alpha coronaviruses including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and human coronavirus, the newly emerged 2019 coronavirus (COVID-19) is considered as a lethal zoonotic virus due to its deadly respiratory syndrome and high mortality rate among the human. Globally, more than 3,517,345 cases have been confirmed with 243,401 deaths due to Acute Respiratory Distress Syndrome (ARDS) caused by COVID-19. The antiviral drug discovery activity is required to control the persistence of COVID-19 circulation and the potential of the future emergence of coronavirus. However, the present review aims to highlight the important antiviral approaches, including interferons, ribavirin, mycophenolic acids, ritonavir, lopinavir, inhibitors, and monoclonal antibodies (mAbs) to provoke the nonstructural proteins and deactivate the structural and essential host elements of the virus to control and treat the infection of COVID-19 by inhibiting the viral entry, viral RNA replication and suppressing the viral protein expression. Moreover, the present review investigates the epidemiology, diagnosis, structure, and replication of COVID-19 for better understanding. It is recommended that these proteases, inhibitors, and antibodies could be a good therapeutic option in drug discovery to control the newly emerged coronavirus.HighlightsCOVID-19 has more than 79.5% identical sequence to SARS-CoV and a 96% identical sequence of the whole genome of bat coronaviruses.Acute respiratory distress syndrome (ARDS), renal failure, and septic shock are the possible clinical symptoms associated with COVID-19.Different antivirals, including interferons, ribavirin, lopinavir, and monoclonal antibodies (mAbs) could be the potent therapeutic agents against COVID-19.The initial clinical trials on hydroquinone in combination with azithromycin showed an admirable result in the reduction of COVID-19.The overexpression of inflammation response, cytokine dysregulation, and induction of apoptosis could be an well-organized factors to reduce the pathogenicity of COVID-19.
Collapse
Affiliation(s)
- Abu Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Khalil ur-Rahman
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Ikram-ul- Haq
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Nazish Jahan
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Mumtaz
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Farman
- Department of Chemistry, University of Engineering and Technology, Lahore, Pakistan
| | - Huma Naeem
- Department of Computer Science, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Faheem Abbas
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Naeem
- Department of Biochemistry, Faculty of Sciences, Bahauddin Zakariya University, Multan, Pakistan
| | - Sania Sadiqa
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Saira Bano
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
33
|
Lazarević M, Battaglia G, Jevtić B, Đedović N, Bruno V, Cavalli E, Miljković Đ, Nicoletti F, Momčilović M, Fagone P. Upregulation of Tolerogenic Pathways by the Hydrogen Sulfide Donor GYY4137 and Impaired Expression of H 2S-Producing Enzymes in Multiple Sclerosis. Antioxidants (Basel) 2020; 9:E608. [PMID: 32664399 PMCID: PMC7402185 DOI: 10.3390/antiox9070608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/22/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to examine the in vitro effects of the slow-releasing H2S donor GYY4137 on the immune cells involved in the pathogenesis of the central nervous system (CNS) autoimmune disease, multiple sclerosis (MS). GYY4137 specifically potentiated TGF-β expression and production in dendritic cells and significantly reduced IFN-γ and IL-17 production in the lymph node and spinal cord T cells obtained from mice immunized with CNS antigens. Both the proportion of FoxP3+ regulatory CD4+ T cells in the lymph node cells, and the percentage of IL-17+ CD4+ T cells in the spinal cord cells were reduced upon culturing with GYY4137. Interestingly, the peripheral blood mononuclear cells obtained from the MS patients had a lower expression of the H2S-producing enzyme, 3-mercaptopyruvate-sulfurtransferase (MPST), in comparison to those obtained from healthy donors. A significant inverse correlation between the expression of MPST and several pro-inflammatory factors was also observed. Further studies on the relevance of the observed results for the pathogenesis and therapy of MS are warranted.
Collapse
Affiliation(s)
- Milica Lazarević
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Giuseppe Battaglia
- Department of Physiology and Pharmacology, Sapienza University, Piazzale A. Moro, 5, 00185 Rome, Italy
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, Italy
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Neda Đedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Valeria Bruno
- Department of Physiology and Pharmacology, Sapienza University, Piazzale A. Moro, 5, 00185 Rome, Italy
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, Italy
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Miljana Momčilović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| |
Collapse
|
34
|
Parvathaneni V, Goyal M, Kulkarni NS, Shukla SK, Gupta V. Nanotechnology Based Repositioning of an Anti-Viral Drug for Non-Small Cell Lung Cancer (NSCLC). Pharm Res 2020; 37:123. [DOI: 10.1007/s11095-020-02848-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
|
35
|
Shmakova A, Germini D, Vassetzky Y. HIV-1, HAART and cancer: A complex relationship. Int J Cancer 2020; 146:2666-2679. [PMID: 31603989 DOI: 10.1002/ijc.32730] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
Abstract
HIV infected people are at higher risk of developing cancer, although it is globally diminished in the era of highly active antiretroviral treatment (HAART). Recently, antioncogenic properties of some HAART drugs were discovered. We discuss the role of HAART in the prevention and improvement of treatment outcomes of cancers in HIV-infected people. We describe different trends in HAART-cancer relationships: cancer-predisposing as well as cancer-preventing. We cover the roles of particular drug regimens in cancer prevention. We also describe the causes of cancer treatment with HAART drugs in HIV-negative people, including ongoing clinical studies that may directly point to a possible independent anti-oncogenic activity of HAART drugs. We conclude that despite potent antioncogenic activities of every class of HAART drugs reported in preclinical models, the evidence to date indicates that their independent clinical impact in HIV-infected people is limited. Improved cancer prevention strategies besides HAART are needed to reduce HIV-cancer-related mortality.
Collapse
Affiliation(s)
- Anna Shmakova
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Diego Germini
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
| | - Yegor Vassetzky
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
- Koltzov Institute of Developmental Biology, Moscow, Russia
| |
Collapse
|
36
|
Synthesis and Modeling Studies of Furoxan Coupled Spiro-Isoquinolino Piperidine Derivatives as NO Releasing PDE 5 Inhibitors. Biomedicines 2020; 8:biomedicines8050121. [PMID: 32423159 PMCID: PMC7277557 DOI: 10.3390/biomedicines8050121] [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: 03/08/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) is considered to be one of the most important intracellular messengers that play an active role as neurotransmitter in regulation of various cardiovascular physiological and pathological processes. Nitric oxide (NO) is a major factor in penile erectile function. NO exerts a relaxing action on corpus cavernosum and penile arteries by activating smooth muscle soluble guanylate cyclase and increasing the intracellular concentration of cyclic guanosine monophosphate (cGMP). Phophodiesterase (PDE) inhibitors have potential therapeutic applications. NO hybridization has been found to improve and extend the pharmacological properties of the parental compound. The present study describes the synthesis of novel furoxan coupled spiro-isoquinolino-piperidine derivatives and their smooth muscle relaxant activity. The study reveals that, particularly 10d (1.50 ± 0.6) and 10g (1.65 ± 0.7) are moderate PDE 5 inhibitors as compared to Sidenafil (1.43 ± 0.5). The observed effect was explained by molecular modelling studies on phosphodiesterase.
Collapse
|
37
|
Kang J, Feng D, Yang F, Tian X, Han W, Jia H. Comparison of rapamycin and methylprednisolone for treating inflammatory muscle disease in a murine model of experimental autoimmune myositis. Exp Ther Med 2020; 20:219-226. [PMID: 32536994 PMCID: PMC7291653 DOI: 10.3892/etm.2020.8716] [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: 04/01/2019] [Accepted: 12/09/2019] [Indexed: 01/03/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of autoimmune inflammatory muscle diseases. Rapamycin has been shown to ameliorate inflammation and improve muscle function in a mouse model of experimental autoimmune myositis (EAM). In the present study, the therapeutic effect of rapamycin was compared with methylprednisolone (MP) on EAM. Mice were injected with myosin for 10 days to induce EAM and were subsequently treated with rapamycin (1.5 mg/kg), MP (40 mg/kg) or placebo (DMSO) for 14 days. The rapamycin-treated group exhibited significantly decreased severe inflammation and improved muscle strength compared with the MP-treated group. The plasma transforming growth factor-β (TGF-β) concentration in the rapamycin-treated group was significantly higher compared with the placebo group. However, both treatment groups exhibited significantly lower plasma interleukin-10 levels compared with the placebo group. Moreover, splenic regulatory T cell frequency in both the rapamycin- and MP-treated animals was significantly lower than that in the animals of the placebo group. Rapamycin showed better immune suppressive effects than MP in this model of EAM, and these effects were likely to be mediated by the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Juan Kang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Dongyun Feng
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Feng Yang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaojia Tian
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Wenjuan Han
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Hongge Jia
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China.,Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518034, P.R. China
| |
Collapse
|
38
|
The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxidants (Basel) 2020; 9:antiox9050374. [PMID: 32365852 PMCID: PMC7278755 DOI: 10.3390/antiox9050374] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023] Open
Abstract
Disturbed redox homeostasis represents a hallmark of cancer phenotypes, affecting cellular metabolism and redox signaling. Since reactive oxygen and nitrogen species (ROS/RNS) are involved in regulation of proliferation and apoptosis, they may play a double-faced role in cancer, entailing protumorigenic and tumor-suppressing effects in early and later stages, respectively. In addition, ROS and RNS impact the activity and communication of all tumor constituents, mediating their reprogramming from anti- to protumorigenic phenotypes, and vice versa. An important role in this dichotomic action is played by the variable amounts of O2 in the tumor microenvironment, which dictates the ultimate outcome of the influence of ROS/RNS on carcinogenesis. Moreover, ROS/RNS levels remarkably influence the cancer response to therapy. The relevance of ROS/RNS signaling in solid tumors is witnessed by the emergence of novel targeted treatments of solid tumors with compounds that target ROS/RNS action and production, such as tyrosine kinase inhibitors and monoclonal antibodies, which might contribute to the complexity of redox regulation in cancer. Prospectively, the dual role of ROS/RNS in the different stages of tumorigenesis through different impact on oxidation and nitrosylation may also allow development of tailored diagnostic and therapeutic approaches.
Collapse
|
39
|
Abstract
Prostate cancer is the second most common cause of cancer-related death in men in the USA, but the effect of prostate cancer diagnosis and treatment on men in a sexual minority group, including men who have sex with men and transgender women, is poorly understood. Efforts to study this population are complicated, as cancer registries do not routinely collect information on sexual orientation. As a result, epidemiological data regarding this population have come from small studies that have included disparate rates of prostate cancer screening, diagnosis and treatment. Qualitative studies indicate that prostate cancer is experienced differently by sexual minorities, with distinct health-care needs that arise owing to differences in sexual practices, social support systems and relationships with the medical community. Notably, sexual minorities have been reported to experience poorer health-related quality of life outcomes than heterosexual men, and tend to have less robust social support systems, experience increased psychological distress caused by sexual dysfunction (areas of which are unmeasured after treatment), experience isolation within the health-care system and express increased levels of dissatisfaction with treatment. The incidence of prostate cancer actually seems to be decreased in men from sexual minorities living with HIV, despite there being no differences in screening and treatment, with poor cancer-specific mortality. Although the literature on patients with prostate cancer in men from sexual minority groups has historically been sparse, peer-reviewed research in this area has grown considerably during the past decade and has become an important field of study.
Collapse
|
40
|
Midgley AC, Wei Y, Li Z, Kong D, Zhao Q. Nitric-Oxide-Releasing Biomaterial Regulation of the Stem Cell Microenvironment in Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1805818. [PMID: 31423672 DOI: 10.1002/adma.201805818] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 06/06/2019] [Indexed: 06/10/2023]
Abstract
Stem cell therapy has proven to be an attractive solution for the treatment of degenerative diseases or injury. However, poor cell engraftment and survival within injured tissues limits the successful use of stem cell therapy within the clinical setting. Nitric oxide (NO) is an important signaling molecule involved in various physiological processes. Emerging evidence supports NO's diverse roles in modulating stem cell behavior, including survival, migration, differentiation, and paracrine secretion of proregenerative factors. Thus, there has been a shift in research focus to concentrate efforts on the delivery of therapeutic concentration ranges of NO to the target tissue sites. Combinatory therapies utilizing biomaterials that control NO generation and support stem cell delivery can be holistic and synergistic approaches to significantly improve tissue regeneration. Here, the focus is on recent developments of various therapeutic platforms, engineered to both transport NO and to enhance stem-cell-mediated regeneration of damaged tissues. New and emerging revelations of how the stem cell microenvironment can be regulated by NO-releasing biomaterials are also highlighted.
Collapse
Affiliation(s)
- Adam C Midgley
- Rongxiang Xu Center for Regenerative Life Science, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zongjin Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Rongxiang Xu Center for Regenerative Life Science, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
41
|
Han J, Tang Y, Zhong M, Wu W. Antitumor effects and mechanisms of 1,25(OH)2D3 in the Pfeiffer diffuse large B lymphoma cell line. Mol Med Rep 2019; 20:5064-5074. [PMID: 31638226 PMCID: PMC6854594 DOI: 10.3892/mmr.2019.10756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 08/06/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) represents the most common subtype of non-Hodgkin lymphoma in China. 1,25-Dihydroxyvitamin D3 [1,25(OH)2D3] has been shown to possess significant antitumor potential and is degraded by 25-hydroxyvitamin D-24-hydroxylase (CYP24A1). In the present study, the role of CYP24A1 and autophagy, and their underlying mechanisms in the anticancer effects of 1,25(OH)2D3 in DLBCL cells, were investigated. It was found that the levels of CYP24A1 in DLBCL lymph node tissues were higher than in hyperplasia lymphadenitis tissue. Moreover, the expression of CYP24A1 was positively associated with the Ann Arbor stage and the International Prognostic Index in patients with DLBCL, and negatively associated with the clinical response to treatment. Patients >60 years of age were found to have a higher level of CYP24A1. 1,25(OH)2D3 inhibited the proliferation of the Pfeiffer DLBCL cell line and increased the G1 phase population of Pfeiffer cells. Rapamycin (RAPA) in combination with 1,25(OH)2D3 increased the G1 phase distribution of Pfeiffer cells. Furthermore, RAPA blocked the increase of CYP24A1 and vitamin D receptor (VDR) expression induced by 1,25(OH)2D3. 1,25(OH)2D3 induced the formation of autophagosomes, increased the expression of autophagy related protein light chain (LC)3II/LC3I and reduced the expression of the ubiquitin binding protein P62. In addition, 1,25(OH)2D3 decreased the phosphorylation of AKT and mammalian target of RAPA (mTOR), and downstream targets eukaryotic translation imitation factor 4E-binding protein 1 and ribosomal protein S6 kinase β-1 in Pfeiffer cells. The results from the present study suggested that CYP24A1 may be a novel prognostic indicator for DLBCL. 1,25(OH)2D3 inhibited proliferation and induced autophagy of Pfeiffer cells. In addition, 1,25(OH)2D3 increased the G1 phase population of Pfeiffer cells. These effects may be mediated by inhibition of the AKT/mTOR/PI3K signaling pathway. RAPA increased the cell cycle arrest induced by 1,25(OH)2D3 by blocking the upregulated expression of CYP24A1 and VDR.
Collapse
Affiliation(s)
- Jing Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yonghong Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Meizuo Zhong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Wenlin Wu
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
42
|
Curcio MF, Batista WL, Castro ED, Strumillo ST, Ogata FT, Alkmim W, Brunialti MKC, Salomão R, Turcato G, Diaz RS, Monteiro HP, Janini LMR. Nitric oxide stimulates a PKC-Src-Akt signaling axis which increases human immunodeficiency virus type 1 replication in human T lymphocytes. Nitric Oxide 2019; 93:78-89. [PMID: 31539562 DOI: 10.1016/j.niox.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 08/12/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
Human immunodeficiency virus (HIV) infections are typically accompanied by high levels of secreted inflammatory cytokines and generation of high levels of reactive oxygen species (ROS). To elucidate how HIV-1 alters the cellular redox environment during viral replication, we used human HIV-1 infected CD4+T lymphocytes and uninfected cells as controls. ROS and nitric oxide (NO) generation, antioxidant enzyme activity, protein phosphorylation, and viral and proviral loads were measured at different times (2-36 h post-infection) in the presence and absence of the NO donor S-nitroso-N-acetylpenicillamine (SNAP). HIV-1 infection increased ROS generation and decreased intracellular NO content. Upon infection, we observed increases in copper/zinc superoxide dismutase (SOD1) and glutathione peroxidase (GPx) activities, and a marked decrease in glutathione (GSH) concentration. Exposure of HIV-1 infected CD4+T lymphocytes to SNAP resulted in an increasingly oxidizing intracellular environment, associated with tyrosine nitration and SOD1 inhibition. In addition, SNAP treatment promoted phosphorylation and activation of the host's signaling proteins, PKC, Src kinase and Akt. Inhibition of PKC leads to inhibition of Src kinase strongly suggesting that PKC is the upstream element in this signaling cascade. Changes in the intracellular redox environment after SNAP treatment had an effect on HIV-1 replication as reflected by increases in proviral and viral loads. In the absence or presence of SNAP, we observed a decrease in viral load in infected CD4+T lymphocytes pre-incubated with the PKC inhibitor GF109203X. In conclusion, oxidative/nitrosative stress conditions derived from exposure of HIV-1-infected CD4+T lymphocytes to an exogenous NO source trigger a signaling cascade involving PKC, Src kinase and Akt. Activation of this signaling cascade appears to be critical to the establishment of HIV-1 infection.
Collapse
Affiliation(s)
- Marli F Curcio
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil.
| | - Wagner L Batista
- Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, São Paulo, Brazil; Department of Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Eloísa D Castro
- Department of Biochemistry/Molecular Biology, CTCMol, Universidade Federal de São Paulo, Brazil
| | - Scheilla T Strumillo
- Department of Biochemistry/Molecular Biology, CTCMol, Universidade Federal de São Paulo, Brazil
| | - Fernando T Ogata
- Structural and Functional Ecology of Ecosystems, Universidade Paulista, Sorocaba, Brazil
| | - Wagner Alkmim
- Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milena K C Brunialti
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Reinaldo Salomão
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gilberto Turcato
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ricardo S Diaz
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Hugo P Monteiro
- Department of Biochemistry/Molecular Biology, CTCMol, Universidade Federal de São Paulo, Brazil
| | - Luiz Mário R Janini
- Department of Medicine/Infectious Diseases, Universidade Federal de São Paulo, São Paulo, Brazil; Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
43
|
Tramutola F, Armentano MF, Berti F, Chiummiento L, Lupattelli P, D'Orsi R, Miglionico R, Milella L, Bisaccia F, Funicello M. New heteroaryl carbamates: Synthesis and biological screening in vitro and in mammalian cells of wild-type and mutant HIV-protease inhibitors. Bioorg Med Chem 2019; 27:1863-1870. [DOI: 10.1016/j.bmc.2019.03.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 01/30/2023]
|
44
|
Paskaš S, Krajnović T, Basile MS, Dunđerović D, Cavalli E, Mangano K, Mammana S, Al-Abed Y, Nicoletti F, Mijatović S, Maksimović-Ivanić D. Senescence as a main mechanism of Ritonavir and Ritonavir-NO action against melanoma. Mol Carcinog 2019; 58:1362-1375. [PMID: 30997718 DOI: 10.1002/mc.23020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022]
Abstract
The main focus of this study is exploring the effect and mechanism of two HIV-protease inhibitors: Ritonavir and Ritonavir-nitric oxide (Ritonavir-NO) on in vitro growth of melanoma cell lines. NO modification significantly improved the antitumor potential of Ritonavir, as the IC50 values of Ritonavir-NO were approximately two times lower than IC50 values of the parental compound. Our results showed for the first time, that both compounds induced senescence in primary and metastatic melanoma cell lines. This transformation was manifested as a change in cell morphology, enlargement of nuclei, increased cellular granulation, upregulation of β-galactosidase activity, lipofuscin granules appearance, higher production of reactive oxygen species and persistent inhibition of proliferation. The expression of p53, as one of the key regulators of senescence, was upregulated after 48 hours of Ritonavir-NO treatment only in metastatic B16F10 cells, ranking it as a late-response event. The development of senescent phenotype was consistent with the alteration of the cytoskeleton-as we observed diminished expression of vinculin, α-actin, and β-tubulin. Permanent inhibition of S6 protein by Ritonavir-NO, but not Ritonavir, could be responsible for a stronger antiproliferative potential of the NO-modified compound. Taken together, induction of senescent phenotype may provide an excellent platform for developing therapeutic approaches based on selective killing of senescent cells.
Collapse
Affiliation(s)
- Svetlana Paskaš
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Tamara Krajnović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Maria S Basile
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Duško Dunđerović
- Institute of Pathology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Eugenio Cavalli
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Santa Mammana
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| |
Collapse
|
45
|
Lu Y, Wang X, Dong H, Wang X, Yang P, Han L, Wang Y, Zheng Z, Zhang W, Zhang L. Bioinformatics analysis of microRNA expression between patients with and without latent tuberculosis infections. Exp Ther Med 2019; 17:3977-3988. [PMID: 30988779 PMCID: PMC6447890 DOI: 10.3892/etm.2019.7424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is a globally prevalent infectious disease. The mechanisms of latent TB infection (LTBI) remain to be fully elucidated and may provide novel approaches for diagnosis. As therapeutic targets and molecular diagnostic markers, microRNAs (miRs) have been studied and utilized in various diseases. In the present study, the differentially expressed miRs (DEMs) in LTBI were screened and analyzed to determine the underlying mechanisms and identify potential biomarkers, thereby contributing to the diagnosis of LTBI. The GSE25435 and GSE29190 datasets from Gene Expression Omnibus were selected for analysis. The 2 datasets were analyzed individually using the Bioconductor package to screen the DEMs with specific cut-off criteria [P<0.01 and |log (fold change)|≥1]. Target gene prediction and interaction network construction were performed using Targetscan, the Search Tool for the Retrieval of Interacting Genes and Proteins and Cytoscape individually, and were merged using the latter tool. The hub genes were finally selected based on their degree of connectivity (DC). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using the KEGG and GENCLIP. A total of 144 DEMs were identified from the 2 datasets. By exploring the overlapping miRs in the two datasets, Homo sapiens (hsa)-miR-29a and hsa-miR-15b were identified to be decreased, while hsa-miR-576-5p, hsa-miR-500 and hsa-miR-155 were identified to be upregulated. hsa-miR-500a-3p and hsa-miR-29a-3p, as well as 4 genes, namely cell division cycle (CDC)42, actin α1, skeletal muscle (ACTA1), phosphatase and tensin homolog (PTEN) and fos proto-oncogene (FOS), were selected as the key factors in this regulatory network. A total of 9 signaling pathways, including phosphoinositide-3 kinase (PI3K)/AKT and 11 biological processes, were identified to be associated with LTBI. In conclusion, the present analysis identified hsa-miR-500a-3p and hsa-miR-29a-3p, as well as CDC42, ACTA1, PTEN and FOS, as the most promising biomarkers and therapeutic candidates for LTBI. The PI3K/AKT signaling pathway is the key signaling pathway implicated in LTBI, and an in-depth investigation of the efficiency of PI3K/AKT signaling inhibitors may be used to prevent a chronic state of infection in LTBI.
Collapse
Affiliation(s)
- Yang Lu
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Xinmin Wang
- Department of Urinary Surgery, The First Affiliated Hospital, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Hongchang Dong
- Department of Biochemistry, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Xiaofang Wang
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Pu Yang
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Ling Han
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Yingzi Wang
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Zhihong Zheng
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Wanjiang Zhang
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Le Zhang
- Department of Pathophysiology, The Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| |
Collapse
|
46
|
Lopinavir-NO, a nitric oxide-releasing HIV protease inhibitor, suppresses the growth of melanoma cells in vitro and in vivo. Invest New Drugs 2019; 37:1014-1028. [PMID: 30706336 DOI: 10.1007/s10637-019-00733-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
We generated a nitric oxide (NO)-releasing derivative of the anti-HIV protease inhibitor lopinavir by linking the NO moiety to the parental drug. We investigated the effects of lopinavir and its derivative lopinavir-NO on melanoma cell lines in vitro and in vivo. Lopinavir-NO exhibited a twofold stronger anticancer action than lopinavir in vitro. These results were successfully translated into syngeneic models of melanoma in vivo, where a significant reduction in tumour volume was observed only in animals treated with lopinavir-NO. Both lopinavir and lopinavir-NO inhibited cell proliferation and induced the trans-differentiation of melanoma cells to Schwann-like cells. In melanoma cancer cell lines, both lopinavir and lopinavir-NO induced morphological changes, minor apoptosis and reactive oxygen species (ROS) production. However, caspase activation and autophagy were detected only in B16 cells, indicating a cell line-specific treatment response. Lopinavir-NO released NO intracellularly, and NO neutralization restored cell viability. Treatment with lopinavir-NO induced only a transient activation of Akt and inhibition of P70S6 kinase. The results of this study identify lopinavir-NO as a promising candidate for further clinical trials in melanoma and possibly other solid tumours.
Collapse
|
47
|
Shi L, Ye L, Liu P, Liu D, Ye G, Chen J, Dong Z. Ulinastatin inhibits apoptosis induced by serum deprivation in mesenchymal stem cells. Mol Med Rep 2019; 19:2397-2406. [PMID: 30664153 DOI: 10.3892/mmr.2019.9847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 10/10/2018] [Indexed: 11/06/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have exhibited great potential in the therapy of cardiovascular disease. However, the application of MSCs is hampered by apoptosis, which reduces the number of cells in the host cardiac microenvironment. Ulinastatin (UTI), a broad‑spectrum protease inhibitor that can be purified from human urine, has attracted attention for its protective effects through its immunomodulatory and anti‑inflammatory properties. The present study aimed to evaluate the effects of UTI on serum deprivation‑induced apoptosis of MSCs and investigate its molecular mechanisms. Cell viability was determined by the MTT assay. Apoptosis was assessed by flow cytometric analysis with Annexin V/propidium iodide staining. The protein levels of cleaved caspase‑3, B‑cell lymphoma‑2 (Bcl‑2) family proteins, total‑Akt and phospho‑Akt were evaluated by western blot. The results of the present study demonstrated that UTI exhibited a protective effect in serum deprived MSCs, as indicated by increased cell viability, and a reduction in the rate of apoptosis and caspase‑3 activation. In addition, treatment with UTI significantly decreased the expression levels of Bcl‑2, Bcl‑extra large and Bcl‑associated X protein. Furthermore, activation of the Akt signaling pathway was involved in the UTI‑induced anti‑apoptotic effects. The present findings indicated that UTI is able to promote the survival of MSCs under serum deprivation conditions. The present study may be helpful in improving the therapeutic efficacy of MSC transplantation used to cure chronic ischemic heart disease.
Collapse
Affiliation(s)
- Linhui Shi
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Longqiang Ye
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Panpan Liu
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Danqin Liu
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Gongjie Ye
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Jiahong Chen
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Zhouzhou Dong
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
48
|
Shi Y, Lin H, Cao J, Cui C. Botulinum toxin type A induces protective autophagy in human dermal microvascular endothelial cells exposed to an in vitro model of ischemia/reperfusion injury. Exp Ther Med 2018; 16:4379-4386. [PMID: 30542387 PMCID: PMC6257827 DOI: 10.3892/etm.2018.6741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/29/2018] [Indexed: 11/10/2022] Open
Abstract
Botulinum toxin type A (BTXA) has been reported to increase the survival of ischemic skin flaps; however, the exact mechanism underlying this effect remains unclear and needs to be further established. The present study aimed to elucidate whether autophagy caused by BTXA functions as a protection mechanism and to identify the mechanisms of its regulation by BTXA in human dermal microvascular endothelial cells (HDMECs) subjected to hypoxia/reoxygenation (H/R)-induced injury. HDMECs were harvested from the upper eyelid tissues of female blepharoplasty patients. HDMECs were exposed to BTXA treatment for 12 h and then subjected to hypoxia for 8 h, followed by reoxygenation for 24 h. Chloroquine diphosphate salt (CQ) was used as an autophagy inhibitor. H/R led to extreme injury to the HDMECs as indicated by the rise in the apoptosis rate, which was significantly attenuated by BTXA pretreatment. The outcomes demonstrated that H/R caused autophagy, as evidenced by a higher type II/type I ratio of light chain 3 (LC3), increased expression of Beclin-1 and increased autophagosome formation. BTXA enhanced autophagy and attenuated apoptosis in a dose-dependent manner, whereas CQ attenuated the BTXA antiapoptotic effects and inhibited the formation of autophagolysosomes, which caused clustering of the LC3-II in cells. In conclusion, autophagy promoted by BTXA serves as a potential protective effect on ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yanyu Shi
- Department of Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Huang Lin
- Department of Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jiankun Cao
- Department of Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Chao Cui
- Department of Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
49
|
Lazarević M, Mazzon E, Momčilović M, Basile MS, Colletti G, Petralia MC, Bramanti P, Nicoletti F, Miljković Đ. The H₂S Donor GYY4137 Stimulates Reactive Oxygen Species Generation in BV2 Cells While Suppressing the Secretion of TNF and Nitric Oxide. Molecules 2018; 23:molecules23112966. [PMID: 30441775 PMCID: PMC6278327 DOI: 10.3390/molecules23112966] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 01/11/2023] Open
Abstract
GYY4137 is a hydrogen sulfide (H2S) donor that has been shown to act in an anti-inflammatory manner in vitro and in vivo. Microglial cells are among the major players in immunoinflammatory, degenerative, and neoplastic disorders of the central nervous system, including multiple sclerosis, Parkinson’s disease, Alzheimer’s disease, and glioblastoma multiforme. So far, the effects of GYY4137 on microglial cells have not been thoroughly investigated. In this study, BV2 microglial cells were stimulated with interferon-gamma and lipopolysaccharide and treated with GYY4137. The agent did not influence the viability of BV2 cells in concentrations up to 200 μM. It inhibited tumor necrosis factor but not interleukin-6 production. Expression of CD40 and CD86 were reduced under the influence of the donor. The phagocytic ability of BV2 cells and nitric oxide production were also affected by the agent. Surprisingly, GYY4137 upregulated generation of reactive oxygen species (ROS) by BV2 cells. The effect was mimicked by another H2S donor, Na2S, and it was not reproduced in macrophages. Our results demonstrate that GYY4137 downregulates inflammatory properties of BV2 cells but increases their ability to generate ROS. Further investigation of this unexpected phenomenon is warranted.
Collapse
Affiliation(s)
- Milica Lazarević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy.
| | - Miljana Momčilović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Maria Sofia Basile
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Giuseppe Colletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy.
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
50
|
Mijatović S, Bramanti A, Nicoletti F, Fagone P, Kaluđerović GN, Maksimović-Ivanić D. Naturally occurring compounds in differentiation based therapy of cancer. Biotechnol Adv 2018; 36:1622-1632. [DOI: 10.1016/j.biotechadv.2018.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022]
|