Next Article in Journal
Crystallization and Optical Behaviour of Nanocomposite Sol-Gel TiO2:Ag Films
Previous Article in Journal
Effect of the Total Saponins of Bupleurum chinense DC. Water Extracts Following Ultrafiltration Pretreatment on Macroporous Resin Adsorption
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis and Biological Activity Evaluation of β-Carboline Derivatives Containing Nitrogen Heterocycles

1
Guizhou Engineering Laboratory for Synthetic Drugs, College of Pharmacy, Guizhou University, Guiyang 550025, China
2
State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, Guiyang 550025, China
3
Research Center for Green Chemistry and Ecological Environment Technology, Guizhou Industry Polytechnic College, Guiyang 550008, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2024, 29(21), 5155; https://doi.org/10.3390/molecules29215155
Submission received: 17 September 2024 / Revised: 20 October 2024 / Accepted: 22 October 2024 / Published: 31 October 2024
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
A series of β-carboline derivatives containing nitrogen heterocycles were designed and synthesized. All compounds were screened for their antitumor activity against four human tumor cell lines (A549, K562, PC-3, T47D). Notably, compound N-(4-(morpholinomethyl)phenyl)-2-((5-(1-(3,4,5-trimethoxyphenyl)-9H-pyrido[3,4-b]indol-3-yl)-1,3,4-oxadiazol-2-yl)thio)acetamide (8q) exhibited significant inhibitory activity against PC-3 cells with an IC50 value of 9.86 µM. Importantly, compound 8q effectively suppressed both the proliferation and migration of PC-3 cells. Mechanistic studies revealed that compound 8q induced cell apoptosis and caused the accumulation of reactive oxygen species (ROS), leading to cell cycle arrest in the G0/G1 phase in PC-3 cells.

1. Introduction

The utilization of natural products in the development of novel drugs is frequently favored due to their distinctive molecular frameworks and extensive pharmacological properties [1]. The tricyclic planar pyrido-[3,4-b]indole ring alkaloid β-carboline, which is extracted from the seeds of Peganum harmala L., has been documented to possess diverse pharmacological activities, particularly antitumor effects [2]. Its presence can be found in various locations, including plants, marine organisms and even human tissues [3]. The antitumor activity of β-carboline alkaloids and their derivatives is exerted through various mechanisms, including the inhibition of both topoisomerases I and II [4,5], cyclin-dependent kinases (CDKs) [6,7], IkappaB kinase (IKK) [8], Polo-like kinases (PLKs) [9], monoamine oxidase (MAO) [10,11] and DNA insertion and binding [12,13,14]. The compound under investigation exhibits a range of diverse biological activities in addition to its currently studied antitumor activity, including antibacterial [15], antiviral [16,17], anti-Alzheimer [18], antidiabetic [19], anti-inflammatory [20], anti-adipogenic [21] and antimalarial [22,23] (Figure 1).
The critical role of many heterocyclic compound systems in drug development lies in their ability to bind with other cyclic systems, thereby augmenting their activity and exhibiting potent anticancer effects against various human malignant tumor cell lines [24,25,26,27]. Oxadiazole activities include but are not limited to anticancer [28], antibacterial [29] and antifungal [30]. Due to its diverse biological activities, oxadiazole plays a crucial role in drug development. Zibotentan, an oxadiazole-based hybrid drug, is currently undergoing clinical trials [31]. Additionally, 1,3,4-oxadiazole is partially present in several commercially available drugs such as furamizole, raltegravir and nesapidil [32]. Moreover, the piperazine ring, as of 2014, has been reported to be the fourth most common type of heterocycle in marketed drugs, and it is usually introduced into the terminal part of the drug to improve molecular water solubility [33]. Overall, heterocycles play a crucial role in drug molecules.
The unique spatial structure of natural products has always made them excellent scaffolds for targeting various molecular targets, ensuring high-quality outcomes [34]. β-Carboline has biological activity due to its three parallel rings forming a special planar aromatic ring, which makes it easy to embed into DNA, causing DNA damage or preventing DNA repair [35,36]. The oxadiazole moiety is frequently employed in drug molecular design as a linking bridge, while the β-carboline exhibits poor water solubility [37]. Therefore, piperazine, as an end element to improve molecular water solubility, is introduced into the target molecular compound. In order to explore small molecules with higher antitumor activity in β-carboline derivatives, the Pictet–Spengler reaction to the C1 and C3 positions of β-carboline introduces different functional groups, which can enhance antitumor activity [37,38].
One commonly employed approach involves the derivation and modification of natural products to obtain more potent compounds. Based on the significant role of heterocycles in drug molecules, in this work, we designed, synthesized and evaluated 3-substituted nitrogen-containing heterocyclic β-carboline derivatives using a highly active fragment molecular hybridization method, with the aim of obtaining target compounds with higher activity (Figure 2).

2. Results and Discussion

2.1. Synthesis

The synthetic route of β-carboline derivatives containing nitrogen heterocycles is shown in Scheme 1. Firstly, the existing and widely existing commodity L-tryptophan and different substituted benzaldehydes are reacted through a Pictet–Spengler reaction to obtain three parallel rings, namely key intermediate 2a. Subsequently, intermediate 5a is obtained through the esterification, oxidation and hydrazine hydrolysis of intermediate 2a. Intermediate 6a is obtained through acidification [39,40], and finally, nucleophilic substitution is carried out with chloroacetylpiperazine to obtain the target compounds 7 series and 8 series [41]. Spectra of compounds (1H NMR, 13C NMR, HRMS) of series 7 and 8 are included in the Supplementary Materials (Figures S1–S103).

2.2. Biological Evaluation

2.2.1. In Vitro Antitumor Activity of Target Compounds

To investigate the antitumor activity of β-carboline derivatives, their inhibitory ability against A549 (human lung cancer cell), K562 (human chronic myeloid leukemia cell), PC-3 (human prostate cancer cell), T47D (H]human breast cancer cell) and Hek-293T (human embryonic kidney cells) were evaluated using MTT assay according to previously reported methods [42]. Cells were purchased from the Cell Bank of the Chinese Academy of Sciences (Kunming, China). The initial screening results showed that most compounds only had poor anticancer activity (Table 1 and Table 2). From the activity data results, it could be seen that compounds with substituted R1 groups had significantly improved water solubility, which was the 8 series. This may be due to the appearance of substituent groups causing a change in steric hindrance. Subsequently, the rescreening process focused exclusively on compounds 8g and 8q, which belong to the more soluble 8 series. As shown in Table 3, only compounds 8g and 8q exhibit activity below 10 μM. The following structure–activity relationship is obtained from Table 3: (1) Substitution at the C-1 position is necessary. During the experiment, the water solubility of the 8 series was generally higher than that of the 7 series, possibly due to the change in steric hindrance caused by the introduction of functional groups. (2) The introduction of nitrogen-containing heterocycles is necessary. For example, compounds 8g and 8q, in addition to the oxadiazole bridge, also connected to the piperazine ring and the morpholine ring, respectively. They displayed promising antitumor activity against PC-3 cells, with IC50 values of 9.56 μM and 9.86 μM, respectively. However, the antitumor activity of 8k (substituted with p-fluoroaniline) without either a piperazine ring or a morpholine ring was greatly reduced, and its inhibitory activity on PC-3 was greater than 40%. The results from Table 1 demonstrate that the majority of compounds belonging to series 7 and 8 exhibited a limited persistent impact on normal Hek-293T cells. Under microscopy, compound 8g was observed to precipitate crystals, potentially compromising the accuracy of antitumor activity measurements. Consequently, compound 8q was selected for further investigation, and detailed studies were conducted to elucidate its antitumor mechanisms in PC-3 cells.

2.2.2. Colony-Forming Assay and Wound-Healing Assay

Upon conducting preliminary phenotype experiments, it was objectively observed that compound 8q exhibited inhibitory effects on both the proliferation and migration of PC-3 cells. As evident from the results depicted in Figure 3A, the inhibitory action of compound 8q on cancer cell proliferation occurred in a concentration-dependent manner. Given that cell migration plays a pivotal role in tumor cell invasion [43], to further validate the impact of compound 8q on PC-3 cell motility, a wound healing assay was conducted. The data depicted in Figure 3B demonstrated that treatment with compound 8q resulted in a concentration-dependent inhibition of PC-3 cell migration. Specifically, after 24 h of treatment, the migration rates at various concentrations were as follows: 29.56% at 10 μM, 26.32% at 20 μM, and 19.99% at 30 μM, compared to the control group rate of 46.41%. Notably, the migration rate of Harmine at a concentration of 30 μM was slightly lower, at 21.51%, than the maximum concentration of compound 8q. These findings underscore the potent inhibitory effect of compound 8q on the motility of PC-3 cells.

2.2.3. Cell Apoptosis Analysis and Intracellular ROS Assay

Apoptosis is an important target for antitumor therapy. To explore the antitumor mechanism of compound 8q, cell apoptosis was detected using flow cytometry. As shown in Figure 4A, at a concentration of 10 μM, compound 8q caused a total apoptosis rate of 63.23% in PC-3 cells, which was equivalent to the apoptosis rate of a positive-drug-Harmine (65.96%) treatment at a concentration of 30 μM. Moreover, reactive oxygen species (ROS) play a significant role in cell growth, differentiation and apoptosis. Excessive ROS can cause oxidative damage and lead to cell apoptosis [44]. After treatment with compound 8q at a concentration of 10 μM for 24 h, it was observed that the mean fluorescence intensity (MFI) value increased to 138; in comparison, the negative control exhibited an MFI value of 19.8, while the positive control, Harmine (30 μM), demonstrated an MFI value of 37.9 (Figure 4B). The results showed that compound 8q at low concentrations could cause an increase in ROS in PC-3 cells, leading to cell apoptosis.

2.2.4. Cell Cycle Assay and Western Blot

To analyze the effect of compound 8q on cell cycle, the cycle of PC-3 cells was detected after being treated with compound 8q for 24 h (Figure 5). In the negative control group, the cycle showed a normal distribution, with distributions of 50.6%, 20.8% and 23.6% in G0/G1, S and G2/M phases, respectively. However, in the dosing group, the concentration of compound 8q was 10 μM, 20 μM and 30 μM, and the distribution of the G0/G1 phase gradually increased, with values of 55.66%, 60.83% and 62.03%, respectively. The results indicated that compound 8q arrested the PC-3 cell cycle in the G0/G1 phase. In addition, Cyclin D1, a cell cycle-related protein, is a key regulator of cycle progression, mainly regulating G1/S phase transition. Meanwhile, it is upregulated in various cancers. The Western blot assay result showed that after treating cells with compound 8q at a concentration of 30 μM, the expression of Cyclin D1 was significantly downregulated. In summary, compound 8q can arrest cells in the G0/G1 phase.

3. Conclusions

Inspired by the unique molecular structure and extensive pharmacological activity of β-carboline, a series of β-carboline derivatives containing oxadiazole and piperazine were designed, synthesized and screened for antitumor activity against four human tumor cell lines (A549, K562, PC-3, T47D). It was found that the substitution position and substituent groups of β-carboline derivatives were closely related to biological activity. The literature suggests that the incorporation of appropriate substituents at C-3 can enhance the antitumor activity of the compounds [45]. Ikeda et al. [46] conducted a synthesis of C-3-substituted β-carboline, and subsequently evaluated the antitumor efficacy of all synthesized compounds against HeLa S-3, Sarcoma 180 and 293T cancer cell lines using MTT assay. Most hybrids exhibited IC50 values ranging from 0.032 to 16 μM, indicating significant potency. Therefore, the antitumor activity of β-carboline was investigated by incorporating oxadiazole and piperazine moieties at the C-3 position of β-carboline using active splicing principles. The activity results indicated that most compounds exhibit lower antitumor activity due to their poor water solubility. Among all the target compounds, series 8 compounds containing 3,4,5-trimethoxyphenyl exhibited higher antitumor activity compared to series 7 compounds. These results indicate that the inclusion of 3,4,5-trimethoxyphenyl can enhance the water solubility of this compound series. Notably, only compounds 8g and 8q demonstrated significant antitumor activity. It is worth mentioning that compound 8q has considerable antitumor activity against K562, PC-3 and T47D, with IC50 values of 11.17 µM, 9.86 µM and 9.96 µM, respectively. Therefore, PC-3 cells were chosen for further mechanistic studies. The colony formation and migration assay indicated that compound 8q could inhibit the proliferation and motility of PC-3 in a dose-dependent manner. Exposure of PC-3 cells to 10 μM compound 8q resulted in the induction of apoptosis and ROS accumulation. Meanwhile, compound 8q could arrest the cell cycle in the G0/G1 phase. A Western blot assay demonstrated that compound 8q downregulated the expression of oncoprotein Cyclin D1, confirming that compound 8q arrested it in G0/G1 phase. However, further experimental exploration is needed to identify the target pathway, and the structure needs to be further optimized to improve its water solubility and antitumor activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules29215155/s1, Figures S1–S103: Spectra of compounds (1H NMR, 13C NMR, HRMS). Refs. [47,48] are cited and included in Supplementary Materials.

Author Contributions

Conceptualization, G.W., D.C. and Z.W.; methodology, G.W., W.W., D.C. and Z.W.; software, G.W., L.S., Y.Z. and Z.L.; validation, G.W., B.L. and D.C.; formal analysis, G.W., S.B. and D.C.; investigation, G.W., Z.W. and D.C.; resources, G.W., F.L., C.X. and Y.Z.; data curation, G.W. and D.C.; writing—original draft preparation, G.W. and D.C.; writing—review and editing, G.W. and D.C.; visualization, D.C.; supervision, D.C.; project administration, D.C.; funding acquisition, D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Top Science and Technology Talent Program of Guizhou Education Department (2022075), the National Natural Science Foundation of China (32360689, 22364008, 32260694), Guizhou Provincial Foundation for Excellent Scholars Program (GCC[2023]072), Guizhou Provincial Natural Science Foundation (ZK[2024]100), Guizhou Provincial Young Science and Technology Talents Development Project (KY[2022]145) and the National Key R&D Program of China (2023YFD1400400).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data will be made available on request.

Conflicts of Interest

The authors declare that they have no known competing financial interest or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Liu, W.; Wang, M.; Guo, Z.; He, Y.; Jia, H.; He, J.; Miao, S.; Ding, Y.; Wang, S. Inspired by bis-β-carboline alkaloids: Construction and antitumor evaluation of a novel bis-β-carboline scaffold as potent antitumor agents. Bioorg. Chem. 2023, 133, 106401. [Google Scholar] [CrossRef] [PubMed]
  2. Xu, Z.; Zhao, H.; Zhu, J.; Qian, J.; Tao, W.; Xie, X.; Ji, D.; Chen, S.; Gao, G.; Li, P.; et al. Rational design of β-carboline as an efficient type I/II photosensitizer to enable hypoxia-tolerant chemo-photodynamic therapy. Bioorg. Chem. 2023, 141, 106875. [Google Scholar] [CrossRef]
  3. Ling, Y.; Xu, C.; Luo, L.; Cao, J.; Feng, J.; Xue, Y.; Zhu, Q.; Ju, C.; Li, F.; Zhang, Y.; et al. Novel β-Carboline/Hydroxamic Acid Hybrids Targeting Both Histone Deacetylase and DNA Display High Anticancer Activity via Regulation of the p53 Signaling Pathway. J. Med. Chem. 2015, 58, 9214–9227. [Google Scholar] [CrossRef] [PubMed]
  4. Sathish, M.; Chetan Dushantrao, S.; Nekkanti, S.; Tokala, R.; Thatikonda, S.; Tangella, Y.; Srinivas, G.; Cherukommu, S.; Hari Krishna, N.; Shankaraiah, N.; et al. Synthesis of DNA interactive C3-trans-cinnamide linked β-carboline conjugates as potential cytotoxic and DNA topoisomerase I inhibitors. Bioorgan. Med. Chem. 2018, 26, 4916–4929. [Google Scholar] [CrossRef]
  5. Sathish, M.; Kavitha, B.; Nayak, V.L.; Tangella, Y.; Ajitha, A.; Nekkanti, S.; Alarifi, A.; Shankaraiah, N.; Nagesh, N.; Kamal, A. Synthesis of podophyllotoxin linked β-carboline congeners as potential anticancer agents and DNA topoisomerase II inhibitors. Eur. J. Med. Chem. 2018, 144, 557–571. [Google Scholar] [CrossRef] [PubMed]
  6. Jenkins, P.R.; Wilson, J.; Emmerson, D.; Garcia, M.D.; Smith, M.R.; Gray, S.J.; Britton, R.G.; Mahale, S.; Chaudhuri, B. Design, synthesis and biological evaluation of new tryptamine and tetrahydro-β-carboline-based selective inhibitors of CDK4. Bioorgan. Med. Chem. 2008, 16, 7728–7739. [Google Scholar] [CrossRef]
  7. Ahmad, I.; Fakhri, S.; Khan, H.; Jeandet, P.; Aschner, M.; Yu, Z. Targeting cell cycle by β-carboline alkaloids in vitro: Novel therapeutic prospects for the treatment of cancer. Chem-Biol. Interact. 2020, 330, 109229. [Google Scholar] [CrossRef]
  8. Komulainen, H.; Tuomisto, J.; Airaksinen, M.M.; Kari, I.; Peura, P.; Pollari, L. Tetrahydro-β-carbolines and Corresponding Tryptamines: In Vitro Inhibition of Serotonin, Dopamine and Noradrenaline Uptake in Rat Brain Synaptosomes. Acta Pharmacol. Toxicol. 2009, 46, 299–307. [Google Scholar] [CrossRef]
  9. Han, X.; Zhang, J.; Guo, L.; Cao, R.; Li, Y.; Li, N.; Ma, Q.; Wu, J.; Wang, Y.; Si, S. A Series of Beta-Carboline Derivatives Inhibit the Kinase Activity of PLKs. PLoS ONE 2012, 7, e46546. [Google Scholar] [CrossRef]
  10. Benny, F.; Kumar, S.; Jayan, J.; Abdelgawad, M.A.; Ghoneim, M.M.; Kumar, A.; Manoharan, A.; Susan, R.; Sudevan, S.T.; Mathew, B. Review of β-carboline and its derivatives as selective MAO-A inhibitors. Arch. Pharm. 2023, 356, 2300091. [Google Scholar] [CrossRef]
  11. Herraiz, T. β-Carboline Alkaloids in Soy Sauce and Inhibition of Monoamine Oxidase (MAO). Molecules 2023, 28, 2723. [Google Scholar] [CrossRef] [PubMed]
  12. Lakshmi Manasa, K.; Thatikonda, S.; Sigalapalli, D.K.; Sagar, A.; Kiranmai, G.; Kalle, A.M.; Alvala, M.; Godugu, C.; Nagesh, N.; Nagendra Babu, B. Design and synthesis of β-carboline linked aryl sulfonyl piperazine derivatives: DNA topoisomerase II inhibition with DNA binding and apoptosis inducing ability. Bioorg. Chem. 2020, 101, 103983. [Google Scholar] [CrossRef] [PubMed]
  13. Nafisi, S.; Bonsaii, M.; Maali, P.; Khalilzadeh, M.A.; Manouchehri, F. β-Carboline alkaloids bind DNA. J. Photochem. Photobiol. B Biol. 2010, 100, 84–91. [Google Scholar] [CrossRef] [PubMed]
  14. Giulietti, J.M.; Tate, P.M.; Cai, A.; Cho, B.; Mulcahy, S.P. DNA-binding studies of the natural β-carboline eudistomin U. Bioorg. Med. Chem. Lett. 2016, 26, 4705–4708. [Google Scholar] [CrossRef]
  15. Venkataramana Reddy, P.O.; Hridhay, M.; Nikhil, K.; Khan, S.; Jha, P.N.; Shah, K.; Kumar, D. Synthesis and investigations into the anticancer and antibacterial activity studies of β-carboline chalcones and their bromide salts. Bioorg. Med. Chem. Lett. 2018, 28, 1278–1282. [Google Scholar] [CrossRef]
  16. Wu, Z.; Chen, N.; Tang, Q.; Chen, S.; Zhan, Z.; Zhang, Y.; Wang, G.; Li, Y.; Ye, W.C. β-Carboline Alkaloids from the Seeds of Peganum harmala and Their Anti-HSV-2 Virus Activities. Org. Lett. 2020, 22, 7310–7314. [Google Scholar] [CrossRef]
  17. Yang, Y.; Huang, Y.; Song, H.; Liu, Y.; Wang, L.; Wang, Q. Skeletal modifications of β-carboline alkaloids and their antiviral activity profile. Mol. Divers. 2016, 20, 829–835. [Google Scholar] [CrossRef]
  18. Beato, A.; Gori, A.; Boucherle, B.; Peuchmaur, M.; Haudecoeur, R. β-Carboline as a Privileged Scaffold for Multitarget Strategies in Alzheimer’s Disease Therapy. J. Med. Chem. 2021, 64, 1392–1422. [Google Scholar] [CrossRef]
  19. Xiao, D.; Lu, L.; Liang, B.; Xiong, Z.; Xu, X.; Chen, W. Identification of 1,3,4-oxadiazolyl-containing β-carboline derivatives as novel α-glucosidase inhibitors with antidiabetic activity. Eur. J. Med. Chem. 2023, 261, 115795. [Google Scholar] [CrossRef]
  20. Kumar, S.; Wang, Y.; Chen, P.; Chang, Y.; Kashyap, H.K.; Shen, Y.; Yu, H.; Hwang, T. Design and synthesis of β-carboline and combretastatin derivatives as anti-neutrophilic inflammatory agents. Bioorg. Chem. 2021, 111, 104846. [Google Scholar] [CrossRef]
  21. Baek, S.C.; Nam, K.H.; Yi, S.A.; Jo, M.S.; Lee, K.H.; Lee, Y.H.; Lee, J.; Kim, K.H. Anti-adipogenic Effect of β-Carboline Alkaloids from Garlic (Allium sativum). Foods 2019, 8, 673. [Google Scholar] [CrossRef] [PubMed]
  22. Kamboj, A.; Sihag, B.; Brar, D.S.; Kaur, A.; Salunke, D.B. Structure activity relationship in β-carboline derived anti-malarial agents. Eur. J. Med. Chem. 2021, 221, 113536. [Google Scholar] [CrossRef]
  23. Gorki, V.; Walter, N.S.; Singh, R.; Chauhan, M.; Dhingra, N.; Salunke, D.B.; Kaur, S. β-Carboline Derivatives Tackling Malaria: Biological Evaluation and Docking Analysis. Acs. Omega 2020, 5, 17993–18006. [Google Scholar] [CrossRef]
  24. Asati, V.; Mahapatra, D.K.; Bharti, S.K. Thiazolidine-2,4-diones as multi-targeted scaffold in medicinal chemistry: Potential anticancer agents. Eur. J. Med. Chem. 2014, 87, 814–833. [Google Scholar] [CrossRef] [PubMed]
  25. Sireesha, R.; Tej, M.B.; Poojith, N.; Sreenivasulu, R.; Musuluri, M.; Subbarao, M. Synthesis of Substituted Aryl Incorporated Oxazolo[4,5-b]Pyridine-Triazole Derivatives: Anticancer Evaluation and Molecular Docking Studies. Polycycl. Aromat. Compd. 2023, 43, 915–932. [Google Scholar] [CrossRef]
  26. Lingam, J.; Sahoo, B.K.; Mallavarapu, B.D.; Sreenivasulu, R. Design, synthesis, anticancer evaluation and molecular docking studies of 1,2,4-oxadiazole incorporated indazole-isoxazole derivatives. Synth. Commun. 2024, 54, 66–76. [Google Scholar] [CrossRef]
  27. Hatti, I.; Sujana, O.; Nookaraju, M.; Suresh, J.; Aparna Seetharam, K.; Raju, R.R. Rational design, synthesis, and anticancer evaluation of pyridine and substituted aryl linked 1,3,4-oxadiazole derivatives. Results Chem. 2024, 11, 101755. [Google Scholar] [CrossRef]
  28. Benassi, A.; Doria, F.; Pirota, V. Groundbreaking Anticancer Activity of Highly Diversified Oxadiazole Scaffolds. Int. J. Mol. Sci. 2020, 21, 8692. [Google Scholar] [CrossRef]
  29. Tian, K.; Li, X.; Zhang, L.; Gan, Y.; Meng, J.; Wu, S.; Wan, J.; Xu, Y.; Cai, C.; Ouyang, G.; et al. Synthesis of novel indole derivatives containing double 1,3,4-oxadiazole moiety as efficient bactericides against phytopathogenic bacterium Xanthomonas oryzae. Chem. Pap. 2019, 73, 17–25. [Google Scholar] [CrossRef]
  30. Rodrigues-Vendramini, F.A.V.; Faria, D.R.; Arita, G.S.; Capoci, I.R.G.; Sakita, K.M.; Caparroz-Assef, S.M.; Becker, T.C.A.; de Souza Bonfim-Mendonça, P.; Felipe, M.S.; Svidzinski, T.I.E.; et al. Antifungal activity of two oxadiazole compounds for the paracoccidioidomycosis treatment. PLoS Negl. Trop. Dis. 2019, 13, e7441. [Google Scholar] [CrossRef]
  31. Nayak, S.; Gaonkar, S.L.; Musad, E.A.; Dawsar, A.M.A. 1,3,4-Oxadiazole-containing hybrids as potential anticancer agents: Recent developments, mechanism of action and structure-activity relationships. J. Saudi Chem. Soc. 2021, 25, 101284. [Google Scholar] [CrossRef]
  32. Paruch, K.; Popiołek, A.; Wujec, M. Antimicrobial and antiprotozoal activity of 3-acetyl-2,5-disubstituted-1,3,4-oxadiazolines: A review. Med. Chem. Res. 2020, 29, 1–16. [Google Scholar] [CrossRef]
  33. Meanwell, N.A.; Loiseleur, O. Applications of Isosteres of Piperazine in the Design of Biologically Active Compounds: Part 2. J. Agric. Food Chem. 2022, 70, 10972–11004. [Google Scholar] [CrossRef] [PubMed]
  34. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  35. Soni, J.P.; Devi, P.; Chemitikanti, S.; Sharma, A.; Swamy, C.V.D.; Phanindranath, R.; Sathish, M.; Nagesh, N.; Godugu, C.; Shankaraiah, N. Design, synthesis and in vitro cytotoxic evaluation of β-carboline tethered quinoline-4-carboxamide conjugates as DNA-interactive Topo II inhibitors. J. Mol. Struct. 2023, 1291, 136001. [Google Scholar] [CrossRef]
  36. Hu, W.; Huang, X.; Wu, J.; Yang, L.; Zheng, Y.; Shen, Y.; Li, Z.; Li, X. Discovery of Novel Topoisomerase II Inhibitors by Medicinal Chemistry Approaches. J. Med. Chem. 2018, 61, 8947–8980. [Google Scholar] [CrossRef]
  37. Ao, J.; Zeng, F.; Wang, L.; Qiu, L.; Cao, R.; Li, X. Design, synthesis and pharmacological evaluation of β-carboline derivatives as potential antitumor agent via targeting autophagy. Eur. J. Med. Chem. 2023, 246, 114955. [Google Scholar] [CrossRef]
  38. Szabó, T.; Volk, B.; Milen, M. Recent Advances in the Synthesis of β-Carboline Alkaloids. Molecules 2021, 26, 663. [Google Scholar] [CrossRef]
  39. Zhang, Z.; Zeng, Y.; Jiang, Z.; Shu, B.; Sethuraman, V.; Zhong, G. Design, synthesis, fungicidal property and QSAR studies of novel β-carbolines containing urea, benzoylthiourea and benzoylurea for the control of rice sheath blight. Pest Manag. Sci. 2018, 74, 1736–1746. [Google Scholar] [CrossRef]
  40. Yao, K.; Zhao, M.; Zhang, X.; Wang, Y.; Li, L.; Zheng, M.; Peng, S. A class of oral N-[(1S,3S)-1-methyl-1,2,3,4-tetrahydro-β-carboline-3-carbonyl]- N′-(amino-acid-acyl)hydrazine: Discovery, synthesis, in vitro anti-platelet aggregation/in vivo anti-thrombotic evaluation and 3D QSAR analysis. Eur. J. Med. Chem. 2011, 46, 3237–3249. [Google Scholar] [CrossRef]
  41. Patel, N.B.; Purohit, A.C.; Rajani, D.P.; Moo-Puc, R.; Rivera, G. New 2-benzylsulfanyl-nicotinic acid based 1,3,4-oxadiazoles: Their synthesis and biological evaluation. Eur. J. Med. Chem. 2013, 62, 677–687. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, C.; Zhu, M.; Long, X.; Wang, Q.; Wang, Z.; Ouyang, G. Design, Synthesis and Antitumor Activity of 1H-indazole-3-amine Derivatives. Int. J. Mol. Sci. 2023, 24, 8686. [Google Scholar] [CrossRef]
  43. Wang, W.; Qin, J.; Voruganti, S.; Srivenugopal, K.S.; Nag, S.; Patil, S.; Sharma, H.; Wang, M.; Wang, H.; Buolamwini, J.K.; et al. The pyridoindole MDM2 inhibitor SP-141 exerts potent therapeutic effects in breast cancer models. Nat. Commun. 2014, 5, 5086. [Google Scholar] [CrossRef] [PubMed]
  44. Zaidieh, T.; Smith, J.R.; Ball, K.E.; An, Q. ROS as a novel indicator to predict anticancer drug efficacy. BMC Cancer 2019, 19, 1224. [Google Scholar] [CrossRef]
  45. Luo, B.; Song, X. A comprehensive overview of beta-carbolines and its derivatives as anticancer agents. Eur. J. Med. Chem. 2021, 224, 113688. [Google Scholar] [CrossRef]
  46. Ikeda, R.; Kimura, T.; Tsutsumi, T.; Tamura, S.; Sakai, N.; Konakahara, T. Structure-activity relationship in the antitumor activity of 6-, 8- or 6,8-substituted 3-benzylamino-beta-carboline derivatives. Bioorg. Med. Chem. Lett. 2012, 22, 3506–3515. [Google Scholar] [CrossRef] [PubMed]
  47. Zou, Y.; Zhang, G.; Li, C.; Long, H.; Chen, D.; Li, Z.; Ouyang, G.; Zhang, W.; Zhang, Y.; Wang, Z. Discovery of Tryptanthrin and Its Derivatives and Its Activities against NSCLC In Vitro via Both Apoptosis and Autophagy Pathways. Int. J. Mol. Sci. 2023, 24, 1450. [Google Scholar] [CrossRef]
  48. Zhang, G.; Tang, Z.; Fan, S.L., C.; Li, Y.; Liu, W.; Long, X.; Zhang, W.; Zhang, Y.; Li, Z.; et al. Synthesis and biological assessment of indole derivatives containing penta-heterocycles scaffold as novel anticancer agents towards A549 and K562 cells. J. Enzyme Inhib. Med. Chem. 2023, 38, 2163393. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of β-carboline derivatives with multiple activities.
Figure 1. Chemical structures of β-carboline derivatives with multiple activities.
Molecules 29 05155 g001
Figure 2. The design concept of β-carboline derivatives.
Figure 2. The design concept of β-carboline derivatives.
Molecules 29 05155 g002
Scheme 1. Synthetic route of target compounds 7 and 8 series.
Scheme 1. Synthetic route of target compounds 7 and 8 series.
Molecules 29 05155 sch001
Figure 3. (A) Compound 8q inhibited the colony formation of PC-3 cells (scale bar: 100 μm). (B) The effect of compound 8q at 10 μM, 20 μM and 30 μM concentration and Harmine at 30 μM concentration on the migration of PC-3 cells. (C) Quantitative analysis of PC-3 cells’ migration rate using Image J software (2.1.0/1.53c). One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05, ** p < 0.01, *** p < 0.001).
Figure 3. (A) Compound 8q inhibited the colony formation of PC-3 cells (scale bar: 100 μm). (B) The effect of compound 8q at 10 μM, 20 μM and 30 μM concentration and Harmine at 30 μM concentration on the migration of PC-3 cells. (C) Quantitative analysis of PC-3 cells’ migration rate using Image J software (2.1.0/1.53c). One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05, ** p < 0.01, *** p < 0.001).
Molecules 29 05155 g003
Figure 4. (A) Compound 8q at 10 μM and Harmine at 30 μM concentrations induced apoptosis of PC-3 cells. (B) Quantitative analysis of PC-3 cell apoptosis using Flow Jo-V10 software. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (**** p < 0.0001). (C) Compound 8q at 10 μM concentration and Harmine at 30 μM increased the level of reactive oxygen species in PC-3 cells. (D) Quantitative analysis of reactive oxygen species in PC-3 cells using Flow Jo-V10 software. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05, **** p < 0.001).
Figure 4. (A) Compound 8q at 10 μM and Harmine at 30 μM concentrations induced apoptosis of PC-3 cells. (B) Quantitative analysis of PC-3 cell apoptosis using Flow Jo-V10 software. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (**** p < 0.0001). (C) Compound 8q at 10 μM concentration and Harmine at 30 μM increased the level of reactive oxygen species in PC-3 cells. (D) Quantitative analysis of reactive oxygen species in PC-3 cells using Flow Jo-V10 software. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05, **** p < 0.001).
Molecules 29 05155 g004
Figure 5. (A) Flow cytometry detection of the effect of compound 8q on the distribution of PC-3 cell cycle at concentrations of 10 μM, 20 μM and 30 μM. (B) Quantitative analysis of cell cycles in PC-3 cells using Flow Jo-V10 software. β-Tubulin as an internal reference. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9. (C) Western blot analysis of the effect of 8q on the expression of Cyclin D1 in cells. (D) Analyzed protein grayscale values using Image J software (2.1.0/1.53c). β-Tubulin as an internal reference. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05).
Figure 5. (A) Flow cytometry detection of the effect of compound 8q on the distribution of PC-3 cell cycle at concentrations of 10 μM, 20 μM and 30 μM. (B) Quantitative analysis of cell cycles in PC-3 cells using Flow Jo-V10 software. β-Tubulin as an internal reference. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9. (C) Western blot analysis of the effect of 8q on the expression of Cyclin D1 in cells. (D) Analyzed protein grayscale values using Image J software (2.1.0/1.53c). β-Tubulin as an internal reference. One-way ANOVA followed by Dunnett’s multiple-comparisons test was performed using GraphPad Prism 9 (* p < 0.05).
Molecules 29 05155 g005
Table 1. In vitro cytotoxicity (10 μM) of the compound 7 series.
Table 1. In vitro cytotoxicity (10 μM) of the compound 7 series.
Compds.Inhibitory Activity a/% at 10 μM
PC-3A549K562T47DHek-293T
7a29.58 ± 0.250.72 ± 1.630.85 ± 1.0132.30 ± 3.3618.56 ± 0.71
7b29.09 ± 0.2713.61 ± 7.888.98 ± 3.1342.62 ± 1.6533.29 ± 0.80
7c22.69 ± 2.1916.75 ± 2.3918.08 ± 2.6340.21 ± 0.9047.29 ± 4.59
7d26.47 ± 5.7611.24 ± 0.6917.82 ± 3.9725.41 ± 4.8236.90 ± 1.74
7e24.97 ± 4.0617.98 ± 2.0612.65 ± 4.7247.69 ± 3.3626.36 ± 1.35
7f11.54 ± 2.7813.54 ± 6.9721.49 ± 3.0242.35 ± 4.2356.84 ± 2.21
7g14.81 ± 2.23/ c0.05 ± 2.9942.52 ± 3.1934.11 ± 6.80
7h10.65 ± 2.682.16 ± 3.568.23 ± 4.1246.42 ± 2.1247.51 ± 0.78
7i35.65 ± 1.895.20 ± 7.2123.60 ± 3.5954.25 ± 3.8884.14 ± 0.79
7j14.72 ± 4.4934.04 ± 0.8713.61 ± 1.2139.88 ± 3.5013.44 ± 0.68
7k35.84 ± 4.0910.11 ± 6.1912.24 ± 0.1847.52 ± 1.3930.77 ± 2.59
7l39.56 ± 5.0612.23 ± 6.149.28 ± 2.5119.32 ± 5.0311.20 ± 7.91
7m20.01 ± 3.2214.72 ± 3.6432.99 ± 1.8325.16 ± 4.1029.48 ± 3.33
7n36.00 ± 3.688.72 ± 2.5735.08 ± 3.1247.75 ± 0.8261.79 ± 1.57
7o5.36 ± 3.63/1.26 ± 3.6625.01 ± 1.4335.33 ± 3.81
7p51.25 ± 3.1117.00 ± 2.108.68 ± 6.8350.69 ± 5.7961.48 ± 3.24
7q29.50 ± 1.388.67 ± 0.6417.01 ± 1.8646.42 ± 2.8632.30 ± 2.21
7r43.98 ± 2.623.03 ± 5.8317.22 ± 2.3538.40 ± 5.850.74 ± 3.77
Harmine b14.46 ± 1.4230.99 ± 2.8136.02 ± 2.7751.28 ± 4.5829.10 ± 3.19
5-Fluorouracild40.58 ± 2.04
Gefitinib37.06 ± 0.50
a Average of three replicates. b Harmine used as a positive control. c “/” means no anticancer activity. d indicates the unmeasured values.
Table 2. In vitro cytotoxicity (10 μM) of the compound 8 series.
Table 2. In vitro cytotoxicity (10 μM) of the compound 8 series.
Compds.Inhibitory Activity a/% at 10 μM
PC-3A549K562T47DHek-293T
8a7.73 ± 1.337.02 ± 4.0221.02 ± 1.657.49 ± 3.2529.40 ± 2.16
8b8.91 ± 5.345.03 ± 5.4210.85 ± 2.3210.64 ± 1.1638.35 ± 3.85
8d17.86 ± 2.8010.08 ± 4.448.62 ± 5.59/ c36.92 ± 2.63
8e26.85 ± 4.3926.77 ± 2.6054.35 ± 2.32/23.36 ± 3.77
8f34.50 ± 0.6813.23 ± 0.567.87 ± 5.826.91 ± 5.1320.21 ± 4.34
8g45.44 ± 0.8235.75 ± 1.176.38 ± 0.5240.65 ± 4.4828.96 ± 4.56
8h12.21 ± 1.1317.88 ± 2.912.46 ± 1.78/29.56 ± 3.04
8i21.31 ± 4.8018.41 ± 1.8025.50 ± 2.720.56 ± 3.3825.55 ± 3.44
8j15.74 ± 3.930.13 ± 4.6224.96 ± 3.95/20.37 ± 1.51
8k44.72 ± 1.8233.22 ± 0.4533.46 ± 8.796.94 ± 3.3829.62 ± 2.33
8m46.58 ± 0.352.70 ± 1.7830.19 ± 2.16/20.97 ± 2.61
8n31.96 ± 1.177.62 ± 3.8438.23 ± 3.7325.64 ± 3.7935.54 ± 1.75
8p19.12 ± 2.798.84 ± 0.3644.59 ± 4.1054.16 ± 4.8728.93 ± 3.22
8q46.07 ± 0.7432.86 ± 4.3025.65 ± 14.8914.53 ± 7.4354.76 ± 0.99
8s41.45 ± 0.1910.72 ± 5.297.59 ± 2.67/60.65 ± 5.81
Harmine b14.46 ± 1.4231.23 ± 2.6736.02 ± 2.7748.76 ± 2.3729.10 ± 3.19
5-Fluorouracild40.58 ± 2.04
Gefitinib37.06 ± 0.50
a Average of three replicates. b Harmine used as a positive control. c “/” means no anticancer activity. d indicates the unmeasured values.
Table 3. In vitro cytotoxicity (IC50) of the compounds.
Table 3. In vitro cytotoxicity (IC50) of the compounds.
Compds.IC50 a/(Mean ± S.D., μM)
PC-3A549K562T47D
8g9.56 ± 1.43>40>40>40
8q9.86 ± 1.0326.18 ± 1.5111.71 ± 0.219.96 ± 0.63
Harmine b24.43 ± 4.4525.92 ± 4.7815.68 ± 2.2738.45 ± 1.17
a Average of three replicates. b Harmine used as a positive control.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, G.; Wang, W.; Li, F.; Xu, C.; Zhou, Y.; Li, Z.; Liu, B.; Shao, L.; Chen, D.; Bai, S.; et al. Design, Synthesis and Biological Activity Evaluation of β-Carboline Derivatives Containing Nitrogen Heterocycles. Molecules 2024, 29, 5155. https://doi.org/10.3390/molecules29215155

AMA Style

Wu G, Wang W, Li F, Xu C, Zhou Y, Li Z, Liu B, Shao L, Chen D, Bai S, et al. Design, Synthesis and Biological Activity Evaluation of β-Carboline Derivatives Containing Nitrogen Heterocycles. Molecules. 2024; 29(21):5155. https://doi.org/10.3390/molecules29215155

Chicago/Turabian Style

Wu, Guiyun, Wenhang Wang, Fulian Li, Chenlu Xu, Yue Zhou, Zhurui Li, Bingqian Liu, Lihui Shao, Danping Chen, Song Bai, and et al. 2024. "Design, Synthesis and Biological Activity Evaluation of β-Carboline Derivatives Containing Nitrogen Heterocycles" Molecules 29, no. 21: 5155. https://doi.org/10.3390/molecules29215155

APA Style

Wu, G., Wang, W., Li, F., Xu, C., Zhou, Y., Li, Z., Liu, B., Shao, L., Chen, D., Bai, S., & Wang, Z. (2024). Design, Synthesis and Biological Activity Evaluation of β-Carboline Derivatives Containing Nitrogen Heterocycles. Molecules, 29(21), 5155. https://doi.org/10.3390/molecules29215155

Article Metrics

Back to TopTop