Hostname: page-component-76dd75c94c-5fx6p Total loading time: 0 Render date: 2024-04-30T09:17:10.011Z Has data issue: false hasContentIssue false

Relapse risk revealed by degree centrality and cluster analysis in heroin addicts undergoing methadone maintenance treatment

Published online by Cambridge University Press:  27 October 2021

Lei Wang
Affiliation:
Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China Department of Nuclear Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Feng Hu
Affiliation:
Department of Radiology, The Hospital of Shaanxi Provincial Geology and Mineral Resources Bureau, Xi'an, P.R. China
Wei Li
Affiliation:
Department of Radiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Qiang Li
Affiliation:
Department of Radiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Yongbin Li
Affiliation:
Department of Radiology, The Second Hospital of Xi'an Medical University, Xi'an, P.R. China
Jia Zhu
Affiliation:
Department of Radiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Xuan Wei
Affiliation:
Department of Radiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Jian Yang
Affiliation:
Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
Jianxin Guo
Affiliation:
Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
Yue Qin
Affiliation:
Department of Radiology, Xi'an Daxing Hospital, Xi'an, P.R. China
Hong Shi
Affiliation:
Department of Radiology, Xi'an No.1 Hospital, Xi'an, P.R. China
Wei Wang*
Affiliation:
Department of Radiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, P.R. China
Yarong Wang*
Affiliation:
Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
*
Authors for correspondence: Yarong Wang, E-mail: wangyr9t@xjtu.edu.cn; Wei Wang, E-mail: m_d_wang@hotmail.com
Authors for correspondence: Yarong Wang, E-mail: wangyr9t@xjtu.edu.cn; Wei Wang, E-mail: m_d_wang@hotmail.com

Abstract

Background

Based on hubs of neural circuits associated with addiction and their degree centrality (DC), this study aimed to construct the addiction-related brain networks for patients diagnosed with heroin dependence undertaking stable methadone maintenance treatment (MMT) and further prospectively identify the ones at high risk for relapse with cluster analysis.

Methods

Sixty-two male MMT patients and 30 matched healthy controls (HC) underwent brain resting-state functional MRI data acquisition. The patients received 26-month follow-up for the monthly illegal-drug-use information. Ten addiction-related hubs were chosen to construct a user-defined network for the patients. Then the networks were discriminated with K-means-clustering-algorithm into different groups and followed by comparative analysis to the groups and HC. Regression analysis was used to investigate the brain regions significantly contributed to relapse.

Results

Sixty MMT patients were classified into two groups according to their brain-network patterns calculated by the best clustering-number-K. The two groups had no difference in the demographic, psychological indicators and clinical information except relapse rate and total heroin consumption. The group with high-relapse had a wider range of DC changes in the cortical−striatal−thalamic circuit relative to HC and a reduced DC in the mesocorticolimbic circuit relative to the low-relapse group. DC activity in NAc, vACC, hippocampus and amygdala were closely related with relapse.

Conclusion

MMT patients can be identified and classified into two subgroups with significantly different relapse rates by defining distinct brain-network patterns even if we are blind to their relapse outcomes in advance. This may provide a new strategy to optimize MMT.

Type
Original Article
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

*

These two authors contributed to this work equally.

References

Alia-Klein, N., Parvaz, M. A., Woicik, P. A., Konova, A. B., Maloney, T., Shumay, E., … Goldstein, R. Z. (2011). Gene x disease interaction on orbitofrontal gray matter in cocaine addiction. Archives of General Psychiatry, 68(3), 283294. doi: 10.1001/archgenpsychiatry.2011.10CrossRefGoogle Scholar
Alvandi, M. S., Bourmpoula, M., Homberg, J. R., & Fathollahi, Y. (2017). Association of contextual cues with morphine reward increases neural and synaptic plasticity in the ventral hippocampus of rats. Addiction Biology, 22(6), 18831894. doi: 10.1111/adb.12547CrossRefGoogle ScholarPubMed
Ambroggi, F., Ishikawa, A., Fields, H. L., & Nicola, S. M. (2008). Basolateral amygdala neurons facilitate reward-seeking behavior by exciting nucleus accumbens neurons. Neuron, 59(4), 648661. doi: 10.1016/j.neuron.2008.07.004CrossRefGoogle ScholarPubMed
Andersen, S. L. (2019). Stress, sensitive periods, and substance abuse. Neurobiology of Stress, 10, 100140. doi: 10.1016/j.ynstr.2018.100140CrossRefGoogle ScholarPubMed
Bale, T. L., & Vale, W. W. (2004). CRF and CRF receptors: Role in stress responsivity and other behaviors. Annual Review of Pharmacology and Toxicology, 44, 525557. doi: 10.1146/annurev.pharmtox.44.101802.121410CrossRefGoogle ScholarPubMed
Bayer, R., Franke, H., Ficker, C., Richter, M., Lessig, R., Buttner, A., & Weber, M. (2015). Alterations of neuronal precursor cells in stages of human adult neurogenesis in heroin addicts. Drug and Alcohol Dependence, 156, 139149. doi: 10.1016/j.drugalcdep.2015.09.005CrossRefGoogle ScholarPubMed
Bickel, W. K., Jarmolowicz, D. P., Mueller, E. T., Gatchalian, K. M., & McClure, S. M. (2012). Are executive function and impulsivity antipodes? A conceptual reconstruction with special reference to addiction. Psychopharmacology, 221(3), 361387. doi: 10.1007/s00213-012-2689-xCrossRefGoogle ScholarPubMed
Borjkhani, M., Bahrami, F., & Janahmadi, M. (2018). Computational modeling of opioid-induced synaptic plasticity in hippocampus. PloS One, 13(3), e0193410. doi: 10.1371/journal.pone.0193410CrossRefGoogle ScholarPubMed
Britt, J. P., Benaliouad, F., McDevitt, R. A., Stuber, G. D., Wise, R. A., & Bonci, A. (2012). Synaptic and behavioral profile of multiple glutamatergic inputs to the nucleus accumbens. Neuron, 76(4), 790803. doi: 10.1016/j.neuron.2012.09.040CrossRefGoogle ScholarPubMed
Buckner, R. L., Sepulcre, J., Talukdar, T., Krienen, F. M., Liu, H., Hedden, T., … Johnson, K. A. (2009). Cortical hubs revealed by intrinsic functional connectivity: Mapping, assessment of stability, and relation to Alzheimer's disease. Journal of Neuroscience, 29(6), 18601873. doi: 10.1523/JNEUROSCI.5062-08.2009CrossRefGoogle ScholarPubMed
Camchong, J., Macdonald, A. W III.., Mueller, B. A., Nelson, B., Specker, S., Slaymaker, V., & Lim, K. O. (2014). Changes in resting functional connectivity during abstinence in stimulant use disorder: A preliminary comparison of relapsers and abstainers. Drug and Alcohol Dependence, 139, 145151. doi: 10.1016/j.drugalcdep.2014.03.024CrossRefGoogle ScholarPubMed
Cao, X., Wu, Z., Rou, K., Li, L., Lin, C., Wang, C., … National Working Group on Methadone Maintenance Treatment, P. (2014). Retention and its predictors among methadone maintenance treatment clients in China: A six-year cohort study. Drug and Alcohol Dependence, 145, 8793. doi: 10.1016/j.drugalcdep.2014.09.776CrossRefGoogle Scholar
Chang, H., Li, W., Li, Q., Chen, J., Zhu, J., Ye, J., … Wang, W. (2016). Regional homogeneity changes between heroin relapse and non-relapse patients under methadone maintenance treatment: A resting-state fMRI study. BMC Neurology, 16(1), 145. doi: 10.1186/s12883-016-0659-3CrossRefGoogle ScholarPubMed
Coller, J. K., Barratt, D. T., Dahlen, K., Loennechen, M. H., & Somogyi, A. A. (2006). ABCB1 Genetic variability and methadone dosage requirements in opioid-dependent individuals. Clinical Pharmacology and Therapeutics, 80(6), 682690. doi: 10.1016/j.clpt.2006.09.011CrossRefGoogle ScholarPubMed
Contreras-Rodriguez, O., Albein-Urios, N., Perales, J. C., Martinez-Gonzalez, J. M., Vilar-Lopez, R., Fernandez-Serrano, M. J., … Verdejo-Garcia, A. (2015). Cocaine-specific neuroplasticity in the ventral striatum network is linked to delay discounting and drug relapse. Addiction, 110(12), 19531962. doi: 10.1111/add.13076CrossRefGoogle ScholarPubMed
Crews, F. T., & Vetreno, R. P. (2011). Addiction, adolescence, and innate immune gene induction. Frontiers in Psychiatry, 2, 19. doi: 10.3389/fpsyt.2011.00019CrossRefGoogle ScholarPubMed
Danos, P., Kasper, S., Grunwald, F., Klemm, E., Krappel, C., Broich, K., … Moller, H. J. (1998). Pathological regional cerebral blood flow in opiate-dependent patients during withdrawal: A HMPAO-SPECT study. Neuropsychobiology, 37(4), 194199. doi: 10.1159/000026502CrossRefGoogle ScholarPubMed
Darker, C. D., Ho, J., Kelly, G., Whiston, L., & Barry, J. (2016). Demographic and clinical factors predicting retention in methadone maintenance: Results from an Irish cohort. Irish Journal of Medical Science, 185(2), 433441. doi: 10.1007/s11845-015-1314-5CrossRefGoogle ScholarPubMed
Di Martino, A., Zuo, X. N., Kelly, C., Grzadzinski, R., Mennes, M., Schvarcz, A., … Milham, M. P. (2013). Shared and distinct intrinsic functional network centrality in autism and attention-deficit/hyperactivity disorder. Biological Psychiatry, 74(8), 623632. doi: 10.1016/j.biopsych.2013.02.011CrossRefGoogle ScholarPubMed
Feng, Y. M., Jia, Y. F., Su, L. Y., Wang, D., Lv, L., Xu, L., & Yao, Y. G. (2013). Decreased mitochondrial DNA copy number in the hippocampus and peripheral blood during opiate addiction is mediated by autophagy and can be salvaged by melatonin. Autophagy, 9(9), 13951406. doi: 10.4161/auto.25468CrossRefGoogle ScholarPubMed
Ford, C. P., Mark, G. P., & Williams, J. T. (2006). Properties and opioid inhibition of mesolimbic dopamine neurons vary according to target location. Journal of Neuroscience, 26(10), 27882797. doi: 10.1523/JNEUROSCI.4331-05.2006CrossRefGoogle ScholarPubMed
Forster, S. E., Dickey, M. W., & Forman, S. D. (2018). Regional cerebral blood flow predictors of relapse and resilience in substance use recovery: A coordinate-based meta-analysis of human neuroimaging studies. Drug and Alcohol Dependence, 185, 93105. doi: 10.1016/j.drugalcdep.2017.12.009CrossRefGoogle ScholarPubMed
Francis, T. C., Chaudhury, D., & Lobo, M. K. (2014). Optogenetics: illuminating the neural bases of rodent behavior. Open Access Animal Physiology, 2014(6), 3351. doi: 10.2147/OAAP.S42339Google Scholar
Garcia-Fuster, M. J., Ramos-Miguel, A., Rivero, G., La Harpe, R., Meana, J. J., & Garcia-Sevilla, J. A. (2008). Regulation of the extrinsic and intrinsic apoptotic pathways in the prefrontal cortex of short- and long-term human opiate abusers. Neuroscience, 157(1), 105119. doi: 10.1016/j.neuroscience.2008.09.002CrossRefGoogle Scholar
Gauthier, G., Eibl, J. K., & Marsh, D. C. (2018). Improved treatment-retention for patients receiving methadone dosing within the clinic providing physician and other health services (onsite) versus dosing at community (offsite) pharmacies. Drug and Alcohol Dependence, 191, 15. doi: 10.1016/j.drugalcdep.2018.04.029CrossRefGoogle ScholarPubMed
Giedd, J. N. (2008). The teen brain: Insights from neuroimaging. Journal of Adolescent Health, 42(4), 335343. doi: 10.1016/j.jadohealth.2008.01.007CrossRefGoogle ScholarPubMed
Gitik, M., Srivastava, V., Hodgkinson, C. A., Shen, P. H., Goldman, D., & Meyerhoff, D. J. (2016). Association of Superoxide Dismutase 2 (SOD2) genotype with gray matter volume shrinkage in chronic alcohol users: Replication and further evaluation of an addiction gene panel. International Journal of Neuropsychopharmacology, 19(9), 18. doi: 10.1093/ijnp/pyw033CrossRefGoogle ScholarPubMed
Goode, T. D., & Maren, S. (2019). Common neurocircuitry mediating drug and fear relapse in preclinical models. Psychopharmacology, 236(1), 415437. doi: 10.1007/s00213-018-5024-3CrossRefGoogle ScholarPubMed
Grace, A. A., & Rosenkranz, J. A. (2002). Regulation of conditioned responses of basolateral amygdala neurons. Physiology and Behavior, 77(4-5), 489493. doi: 10.1016/s0031-9384(02)00909-5CrossRefGoogle ScholarPubMed
Guo, Z., Liu, X., Hou, H., Wei, F., Liu, J., & Chen, X. (2016). Abnormal degree centrality in Alzheimer's disease patients with depression: A resting-state functional magnetic resonance imaging study. Experimental Gerontology, 79, 6166. doi: 10.1016/j.exger.2016.03.017CrossRefGoogle ScholarPubMed
Holmes, A. J., Hollinshead, M. O., Roffman, J. L., Smoller, J. W., & Buckner, R. L. (2016). Individual differences in cognitive control circuit anatomy link sensation seeking, impulsivity, and substance use. Journal of Neuroscience, 36(14), 40384049. doi: 10.1523/JNEUROSCI.3206-15.2016CrossRefGoogle ScholarPubMed
Hua, K., Wang, T., Li, C., Li, S., Ma, X., Li, C., … Jiang, G. (2018). Abnormal degree centrality in chronic users of codeine-containing cough syrups: A resting-state functional magnetic resonance imaging study. Neuroimage. Clinical, 19, 775781. doi: 10.1016/j.nicl.2018.06.003CrossRefGoogle ScholarPubMed
Jaremko, K. M., Sterling, R. C., & Van Bockstaele, E. J. (2015). Psychological and physiological stress negatively impacts early engagement and retention of opioid-dependent individuals on methadone maintenance. Journal of Substance Abuse Treatment, 48(1), 117127. doi: 10.1016/j.jsat.2014.08.006CrossRefGoogle ScholarPubMed
Jasinska, A. J., Stein, E. A., Kaiser, J., Naumer, M. J., & Yalachkov, Y. (2014). Factors modulating neural reactivity to drug cues in addiction: A survey of human neuroimaging studies. Neuroscience and Biobehavioral Reviews, 38, 116. doi: 10.1016/j.neubiorev.2013.10.013CrossRefGoogle ScholarPubMed
Jenkinson, M., Beckmann, C. F., Behrens, T. E., Woolrich, M. W., & Smith, S. M. (2012). Fsl. Neuroimage, 62(2), 782790. doi: 10.1016/j.neuroimage.2011.09.015CrossRefGoogle ScholarPubMed
Jiang, G. H., Qiu, Y. W., Zhang, X. L., Han, L. J., Lv, X. F., Li, L. M., … Tian, J. Z. (2011). Amplitude low-frequency oscillation abnormalities in the heroin users: A resting state fMRI study. Neuroimage, 57(1), 149154. doi: 10.1016/j.neuroimage.2011.04.004CrossRefGoogle ScholarPubMed
Jiang, H., Han, Y., Du, J., Wu, F., Zhang, R., Zhang, H., … Zhao, M. (2014). Factors associated with one year retention to methadone maintenance treatment program among patients with heroin dependence in China. Substance Abuse Treatment, Prevention, and Policy, 9, 11. doi: 10.1186/1747-597X-9-11CrossRefGoogle ScholarPubMed
Konova, A. B., Moeller, S. J., & Goldstein, R. Z. (2013). Common and distinct neural targets of treatment: Changing brain function in substance addiction. Neuroscience and Biobehavioral Reviews, 37(10 Pt 2), 28062817. doi: 10.1016/j.neubiorev.2013.10.002CrossRefGoogle ScholarPubMed
Koob, G. F. (2009). Brain stress systems in the amygdala and addiction. Brain Research, 1293, 6175. doi: 10.1016/j.brainres.2009.03.038CrossRefGoogle ScholarPubMed
Koob, G. F., & Volkow, N. D. (2010). Neurocircuitry of Addiction. Neuropsychopharmacology, 35(1), 217238. doi: 10.1038/npp.2009.110CrossRefGoogle ScholarPubMed
Lancaster, J. L., Woldorff, M. G., Parsons, L. M., Liotti, M., Freitas, C. S., Rainey, L., … Fox, P. T. (2000). Automated talairach atlas labels for functional brain mapping. Human Brain Mapping, 10(3), 120131. doi: 10.1002/1097-0193(200007)10:3<120::aid-hbm30>3.0.co;2-83.0.CO;2-8>CrossRefGoogle ScholarPubMed
Li, M., Tian, J., Zhang, R., Qiu, Y., Wen, X., Ma, X., … Huang, R. (2014). Abnormal cortical thickness in heroin-dependent individuals. Neuroimage, 88, 295307. doi: 10.1016/j.neuroimage.2013.10.021CrossRefGoogle ScholarPubMed
Li, N., Wang, J., Wang, X. L., Chang, C. W., Ge, S. N., Gao, L., … Gao, G. D. (2013). Nucleus accumbens surgery for addiction. World Neurosurgery, 80(3–4), S28, e9–19. doi: 10.1016/j.wneu.2012.10.007CrossRefGoogle ScholarPubMed
Li, Q., Li, W., Wang, H., Wang, Y., Zhang, Y., Zhu, J., … Liu, Y. (2015a). Predicting subsequent relapse by drug-related cue-induced brain activation in heroin addiction: An event-related functional magnetic resonance imaging study. Addiction Biology, 20(5), 968978. doi: 10.1111/adb.12182CrossRefGoogle ScholarPubMed
Li, Q., Li, Z., Li, W., Zhang, Y., Wang, Y., Zhu, J., … Liu, Y. (2016a). Disrupted default mode network and basal craving in male heroin-dependent individuals: A resting-state fMRI study. Journal of Clinical Psychiatry, 77(10), e1211e1217. doi: 10.4088/JCP.15m09965CrossRefGoogle ScholarPubMed
Li, Q., Liu, J., Wang, W., Wang, Y., Li, W., Chen, J., … Wang, W. (2018). Disrupted coupling of large-scale networks is associated with relapse behaviour in heroin-dependent men. Journal of Psychiatry and Neuroscience, 43(1), 4857. doi: 10.1503/jpn.170011CrossRefGoogle ScholarPubMed
Li, S., Ma, X., Huang, R., Li, M., Tian, J., Wen, H., … Jiang, G. (2016b). Abnormal degree centrality in neurologically asymptomatic patients with end-stage renal disease: A resting-state fMRI study. Clinical Neurophysiology, 127(1), 602609. doi: 10.1016/j.clinph.2015.06.022CrossRefGoogle ScholarPubMed
Li, W., Li, Q., Wang, D., Xiao, W., Liu, K., Shi, L., … Wang, W. (2015b). Dysfunctional default mode network in methadone treated patients who have a higher heroin relapse risk. Scientific Reports, 5, 15181. doi: 10.1038/srep15181CrossRefGoogle ScholarPubMed
Li, W., Zhu, J., Li, Q., Ye, J., Chen, J., Liu, J., … Wang, W. (2016c). Brain white matter integrity in heroin addicts during methadone maintenance treatment is related to relapse propensity. Brain and Behavior, 6(2), e00436. doi: 10.1002/brb3.436CrossRefGoogle ScholarPubMed
Lin, C. K., Hung, C. C., Peng, C. Y., Chao, E., & Lee, T. S. (2015). Factors associated with methadone treatment duration: A Cox regression analysis. PloS One, 10(4), e0123687. doi: 10.1371/journal.pone.0123687CrossRefGoogle ScholarPubMed
Lister, J. J., Brown, S., Greenwald, M. K., & Ledgerwood, D. M. (2019). Gender-specific predictors of methadone treatment outcomes among African Americans at an urban clinic. Substance Abuse, 40(2), 185193. doi: 10.1080/08897077.2018.1547810CrossRefGoogle ScholarPubMed
Liu, H., Li, L., Hao, Y., Cao, D., Xu, L., Rohrbaugh, R., … Liu, Z. (2008). Disrupted white matter integrity in heroin dependence: A controlled study utilizing diffusion tensor imaging. American Journal of Drug and Alcohol Abuse, 34(5), 562575. doi: 10.1080/00952990802295238CrossRefGoogle ScholarPubMed
Liu, W., Liu, H., Wei, D., Sun, J., Yang, J., Meng, J., … Qiu, J. (2015). Abnormal degree centrality of functional hubs associated with negative coping in older Chinese adults who lost their only child. Biological Psychology, 112, 4655. doi: 10.1016/j.biopsycho.2015.09.005CrossRefGoogle ScholarPubMed
London, E. D., Broussolle, E. P., Links, J. M., Wong, D. F., Cascella, N. G., Dannals, R. F., … Rippetoe, L. R. (1990). Morphine-induced metabolic changes in human brain. Studies with positron emission tomography and [fluorine 18]fluorodeoxyglucose. Archives of General Psychiatry, 47(1), 7381. doi: 10.1001/archpsyc.1990.01810130075010CrossRefGoogle ScholarPubMed
Luigjes, J., van den Brink, W., Feenstra, M., van den Munckhof, P., Schuurman, P. R., Schippers, R., … Denys, D. (2012). Deep brain stimulation in addiction: A review of potential brain targets. Molecular Psychiatry, 17(6), 572583. doi: 10.1038/mp.2011.114CrossRefGoogle ScholarPubMed
Luo, X., Guo, L., Dai, X. J., Wang, Q., Zhu, W., Miao, X., & Gong, H. (2017). Abnormal intrinsic functional hubs in alcohol dependence: Evidence from a voxelwise degree centrality analysis. Neuropsychiatric Disease and Treatment, 13, 20112020. doi: 10.2147/NDT.S142742CrossRefGoogle ScholarPubMed
Maldjian, J. A., Laurienti, P. J., Kraft, R. A., & Burdette, J. H. (2003). An automated method for neuroanatomic and cytoarchitectonic atlas-based interrogation of fMRI data sets. Neuroimage, 19(3), 12331239. doi: 10.1016/s1053-8119(03)00169-1CrossRefGoogle ScholarPubMed
McGlone, L., Mactier, H., Hamilton, R., Bradnam, M. S., Boulton, R., Borland, W., … McCulloch, D. L. (2008). Visual evoked potentials in infants exposed to methadone in utero. Archives of Disease in Childhood, 93(9), 784786. doi: 10.1136/adc.2007.132985CrossRefGoogle ScholarPubMed
Mei, W., Zhang, J. X., & Xiao, Z. (2010). Acute effects of sublingual buprenorphine on brain responses to heroin-related cues in early-abstinent heroin addicts: An uncontrolled trial. Neuroscience, 170(3), 808815. doi: 10.1016/j.neuroscience.2010.07.033CrossRefGoogle ScholarPubMed
Muller, U. J., Truebner, K., Schiltz, K., Kuhn, J., Mawrin, C., Dobrowolny, H., … Steiner, J. (2015). Postmortem volumetric analysis of the nucleus accumbens in male heroin addicts: Implications for deep brain stimulation. European Archives of Psychiatry and Clinical Neuroscience, 265(8), 647653. doi: 10.1007/s00406-015-0617-xCrossRefGoogle ScholarPubMed
Naqvi, N. H., Gaznick, N., Tranel, D., & Bechara, A. (2014). The insula: A critical neural substrate for craving and drug seeking under conflict and risk. Annals of the New York Academy of Sciences, 1316, 5370. doi: 10.1111/nyas.12415CrossRefGoogle ScholarPubMed
Nazari-Serenjeh, F., Rezaee, L., Zarrabian, S., & Haghparast, A. (2018). Comparison of the role of D1- and D2-like receptors in the CA1 region of the hippocampus in the reinstatement induced by a subthreshold dose of morphine and forced swim stress in extinguished morphine-CPP in rats. Neurochemical Research, 43(11), 20922101. doi: 10.1007/s11064-018-2631-7CrossRefGoogle ScholarPubMed
Nestler, E. J. (2014). Epigenetic mechanisms of drug addiction. Neuropharmacology, 76(Pt B), 259268. doi: 10.1016/j.neuropharm.2013.04.004CrossRefGoogle ScholarPubMed
Noble, E. P. (2000). Addiction and its reward process through polymorphisms of the D2 dopamine receptor gene: A review. European Psychiatry, 15(2), 7989. doi: 10.1016/s0924-9338(00)00208-xCrossRefGoogle ScholarPubMed
Oliver, R. J., Brigman, J. L., Bolognani, F., Allan, A. M., Neisewander, J. L., & Perrone-Bizzozero, N. I. (2018). Neuronal RNA-binding protein HuD regulates addiction-related gene expression and behavior. Genes, Brain, and Behavior, 17(4), e12454. doi: 10.1111/gbb.12454CrossRefGoogle ScholarPubMed
Owens, M. M., MacKillop, J., Gray, J. C., Beach, S. R. H., Stein, M. D., Niaura, R. S., … Sweet, L. H. (2017). Neural correlates of tobacco cue reactivity predict duration to lapse and continuous abstinence in smoking cessation treatment. Addiction Biology, 23(5), 11891199. doi: 10.1111/adb.12549CrossRefGoogle ScholarPubMed
Pascoli, V., Terrier, J., Espallergues, J., Valjent, E., O'Connor, E. C., & Luscher, C. (2014). Contrasting forms of cocaine-evoked plasticity control components of relapse. Nature, 509(7501), 459464. doi: 10.1038/nature13257CrossRefGoogle ScholarPubMed
Pirompanich, P., & Chankrachang, S. (2015). Intravenous heroin-associated delayed spongiform leukoencephalopathy: Case report and reviews of the literature. Journal of the Medical Association of Thailand, 98(7), 703708, https://www.ncbi.nlm.nih.gov/pubmed/26267994.Google ScholarPubMed
Power, J. D., Barnes, K. A., Snyder, A. Z., Schlaggar, B. L., & Petersen, S. E. (2012). Spurious but systematic correlations in functional connectivity MRI networks arise from subject motion. Neuroimage, 59(3), 21422154. doi: 10.1016/j.neuroimage.2011.10.018CrossRefGoogle ScholarPubMed
Proctor, S. L., Copeland, A. L., Kopak, A. M., Hoffmann, N. G., Herschman, P. L., & Polukhina, N. (2015). Predictors of patient retention in methadone maintenance treatment. Psychology of Addictive Behaviors, 29(4), 906917. doi: 10.1037/adb0000090CrossRefGoogle ScholarPubMed
Randesi, M., van den Brink, W., Levran, O., Yuferov, V., Blanken, P., van Ree, J. M., … Kreek, M. J. (2018). Dopamine gene variants in opioid addiction: Comparison of dependent patients, nondependent users and healthy controls. Pharmacogenomics, 19(2), 95104. doi: 10.2217/pgs-2017-0134CrossRefGoogle ScholarPubMed
Rothenberg, S., Peck, E. A., Schottenfeld, S., Betley, G. E., & Altman, J. L. (1979). Methadone depression of visual signal detection performance. Pharmacology, Biochemistry and Behavior, 11(5), 521527. doi: 10.1016/0091-3057(79)90035-2CrossRefGoogle ScholarPubMed
See, R. E., Fuchs, R. A., Ledford, C. C., & McLaughlin, J. (2003). Drug addiction, relapse, and the amygdala. Annals of the New York Academy of Sciences, 985, 294307. doi: 10.1111/j.1749-6632.2003.tb07089.xCrossRefGoogle ScholarPubMed
Shapira-Lichter, I., Strauss, I., Oren, N., Gazit, T., Sammartino, F., Giacobbe, P., … Lozano, A. M. (2018). Conflict monitoring mechanism at the single-neuron level in the human ventral anterior cingulate cortex. Neuroimage, 175, 4555. doi: 10.1016/j.neuroimage.2018.03.028CrossRefGoogle ScholarPubMed
Shi, J., Hui, L., Xu, Y., Wang, F., Huang, W., & Hu, G. (2002). Sequence variations in the mu-opioid receptor gene (OPRM1) associated with human addiction to heroin. Human Mutation, 19(4), 459460. doi: 10.1002/humu.9026CrossRefGoogle ScholarPubMed
Smith, S. M., Jenkinson, M., Woolrich, M. W., Beckmann, C. F., Behrens, T. E., Johansen-Berg, H., … Matthews, P. M. (2004). Advances in functional and structural MR image analysis and implementation as FSL. Neuroimage, 23(Suppl 1), S208S219. doi: 10.1016/j.neuroimage.2004.07.051CrossRefGoogle ScholarPubMed
Su, H., Li, Z., Du, J., Jiang, H., Chen, Z., Sun, H., & Zhao, M. (2015). Predictors of heroin relapse: Personality traits, impulsivity, COMT gene Val158met polymorphism in a 5-year prospective study in Shanghai, China. American Journal of Medical Genetics. Part B: Neuropsychiatric Genetics, 168(8), 712719. doi: 10.1002/ajmg.b.32376CrossRefGoogle Scholar
Sun, H., Lui, S., Yao, L., Deng, W., Xiao, Y., Zhang, W., … Gong, Q. (2015). Two patterns of white matter abnormalities in medication-naive patients with first-episode schizophrenia revealed by diffusion tensor imaging and cluster analysis. JAMA Psychiatry, 72(7), 678686. doi: 10.1001/jamapsychiatry.2015.0505CrossRefGoogle ScholarPubMed
Takeuchi, H., Taki, Y., Nouchi, R., Sekiguchi, A., Hashizume, H., Sassa, Y., … Kawashima, R. (2015). Degree centrality and fractional amplitude of low-frequency oscillations associated with stroop interference. Neuroimage, 119, 197209. doi: 10.1016/j.neuroimage.2015.06.058CrossRefGoogle ScholarPubMed
Tramullas, M., Martinez-Cue, C., & Hurle, M. A. (2008). Chronic administration of heroin to mice produces up-regulation of brain apoptosis-related proteins and impairs spatial learning and memory. Neuropharmacology, 54(4), 640652. doi: 10.1016/j.neuropharm.2007.11.018CrossRefGoogle ScholarPubMed
Tsou, T.-S. (2006). Robust Poisson regression. Journal of Statistical Planning and Inference, 136(9), 31733186. doi: 10.1016/j.jspi.2004.12.008CrossRefGoogle Scholar
Vassoler, F. M., White, S. L., Hopkins, T. J., Guercio, L. A., Espallergues, J., Berton, O., … Pierce, R. C. (2013). Deep brain stimulation of the nucleus accumbens shell attenuates cocaine reinstatement through local and antidromic activation. Journal of Neuroscience, 33(36), 1444614454. doi: 10.1523/JNEUROSCI.4804-12.2013CrossRefGoogle ScholarPubMed
Volkow, N., & Li, T. K. (2005). The neuroscience of addiction. Nature Neuroscience, 8(11), 14291430. doi: 10.1038/nn1105-1429CrossRefGoogle ScholarPubMed
Volkow, N. D., Fowler, J. S., & Wang, G. J. (2003). The addicted human brain: Insights from imaging studies. Journal of Clinical Investigation, 111(10), 14441451. doi: 10.1172/JCI18533CrossRefGoogle ScholarPubMed
Volkow, N. D., Jones, E. B., Einstein, E. B., & Wargo, E. M. (2019). Prevention and treatment of opioid misuse and addiction: A review. JAMA Psychiatry, 76(2), 208216. doi: 10.1001/jamapsychiatry.2018.3126CrossRefGoogle ScholarPubMed
Volkow, N. D., & Morales, M. (2015). The brain on drugs: From reward to addiction. Cell, 162(4), 712725. doi: 10.1016/j.cell.2015.07.046CrossRefGoogle ScholarPubMed
Volkow, N. D., Wang, G. J., Fowler, J. S., & Tomasi, D. (2012). Addiction circuitry in the human brain. Annual Review of Pharmacology and Toxicology, 52, 321336. doi: 10.1146/annurev-pharmtox-010611-134625CrossRefGoogle ScholarPubMed
Volkow, N. D., Wang, G. J., Fowler, J. S., Tomasi, D., & Telang, F. (2011). Addiction: Beyond dopamine reward circuitry. Proceedings of the National Academy of Sciences of the United States of America, 108(37), 1503715042. doi: 10.1073/pnas.1010654108CrossRefGoogle ScholarPubMed
Volkow, N. D., Wang, G. J., Tomasi, D., & Baler, R. D. (2013). Unbalanced neuronal circuits in addiction. Current Opinion in Neurobiology, 23(4), 639648. doi: 10.1016/j.conb.2013.01.002CrossRefGoogle ScholarPubMed
Wang, G. Y., Kydd, R. R., & Russell, B. R. (2016). Quantitative EEG and Low-Resolution Electromagnetic Tomography (LORETA) imaging of patients undergoing methadone treatment for opiate addiction. Clinical EEG and Neuroscience, 47(3), 180187. doi: 10.1177/1550059415586705CrossRefGoogle ScholarPubMed
Wang, W., Wang, Y. R., Qin, W., Yuan, K., Tian, J., Li, Q., … Guo, Y. M. (2010). Changes in functional connectivity of ventral anterior cingulate cortex in heroin abusers. Chinese Medical Journal (Engl.), 123(12), 15821588, https://www.ncbi.nlm.nih.gov/pubmed/20819516.Google ScholarPubMed
Wang, X., Jiao, D., Zhang, X., & Lin, X. (2017). Altered degree centrality in childhood absence epilepsy: A resting-state fMRI study. Journal of the Neurological Sciences, 373, 274279. doi: 10.1016/j.jns.2016.12.054CrossRefGoogle ScholarPubMed
Wang, Y., Zhu, J., Li, Q., Li, W., Wu, N., Zheng, Y., … Wang, W. (2013). Altered fronto-striatal and fronto-cerebellar circuits in heroin-dependent individuals: A resting-state FMRI study. PloS One, 8(3), e58098. doi: 10.1371/journal.pone.0058098CrossRefGoogle ScholarPubMed
Wei, X., Wang, L., Wang, X., Li, J., Li, H., & Jia, W. (2013). A study of 6-year retention in methadone maintenance treatment among opioid-dependent patients in Xi'an. Journal of Addiction Medicine, 7(5), 342348. doi: 10.1097/ADM.0b013e31829da05bCrossRefGoogle ScholarPubMed
Wolff, K., Hay, A. W., Raistrick, D., & Calvert, R. (1993). Steady-state pharmacokinetics of methadone in opioid addicts. European Journal of Clinical Pharmacology, 44(2), 189194. doi: 10.1007/BF00315479CrossRefGoogle ScholarPubMed
Wollman, S. C., Alhassoon, O. M., Stern, M. J., Hall, M. G., Rompogren, J., Kimmel, C. L., & Perez-Figueroa, A. M. (2015). White matter abnormalities in long-term heroin users: A preliminary neuroimaging meta-analysis. American Journal of Drug and Alcohol Abuse, 41(2), 133138. doi: 10.3109/00952990.2014.985829CrossRefGoogle ScholarPubMed
Woolrich, M. W., Jbabdi, S., Patenaude, B., Chappell, M., Makni, S., Behrens, T., … Smith, S. M. (2009). Bayesian analysis of neuroimaging data in FSL. Neuroimage, 45(1 Suppl), S173S186. doi: 10.1016/j.neuroimage.2008.10.055CrossRefGoogle ScholarPubMed
Wright, V. L., Georgiou, P., Bailey, A., Heal, D. J., Bailey, C. P., & Wonnacott, S. (2018). Inhibition of alpha7 nicotinic receptors in the ventral hippocampus selectively attenuates reinstatement of morphine-conditioned place preference and associated changes in AMPA receptor binding. Addiction Biology, 24(4), 590603. doi: 10.1111/adb.12624CrossRefGoogle ScholarPubMed
Yang, C. H., Choi, S. H., Kim, J. S., Ryu, Y. H., Lim, Y. J., Kim, M. S., … Lee, M. Y. (2017). The effects of acupuncture stimulation for brain activation and alcohol abstinence self-efficacy: Functional MRI study. Evidence-Based Complementary and Alternative Medicine, 2017, 2850124. doi: 10.1155/2017/2850124Google ScholarPubMed
Ye, J. J., Li, W., Zhang, D. S., Li, Q., Zhu, J., Chen, J. J., … Wang, W. (2018). Longitudinal behavioral and fMRI-based assessment of inhibitory control in heroin addicts on methadone maintenance treatment. Experimental and Therapeutic Medicine, 16(4), 32023210. doi: 10.3892/etm.2018.6571Google ScholarPubMed
Zhang, L., Chow, E. P., Zhuang, X., Liang, Y., Wang, Y., Tang, C., … Wilson, D. P. (2013). Methadone maintenance treatment participant retention and behavioural effectiveness in China: A systematic review and meta-analysis. PloS One, 8(7), e68906. doi: 10.1371/journal.pone.0068906CrossRefGoogle Scholar
Zhang, Y., Li, Q., Wen, X., Cai, W., Li, G., Tian, J., … Wang, G. J. (2017). Granger causality reveals a dominant role of memory circuit in chronic opioid dependence. Addiction Biology, 22(4), 10681080. doi: 10.1111/adb.12390CrossRefGoogle ScholarPubMed
Zhou, K., Li, H., Wei, X., Li, X., & Zhuang, G. (2017). Relationships between perceived social support and retention among patients in methadone maintenance treatment in mainland China. Psychology. Health & Medicine, 22(4), 493500. doi: 10.1080/13548506.2016.1164873Google Scholar
Zhou, K., & Zhuang, G. (2014). Retention in methadone maintenance treatment in mainland China, 2004-2012: A literature review. Addictive Behaviors, 39(1), 2229. doi: 10.1016/j.addbeh.2013.09.001CrossRefGoogle ScholarPubMed
Zhou, Y., Wang, Y., Rao, L. L., Liang, Z. Y., Chen, X. P., Zheng, D., … Li, S. (2014). Disrutpted resting-state functional architecture of the brain after 45-day simulated microgravity. Frontiers in Behavioral Neuroscience, 8, 200. doi: 10.3389/fnbeh.2014.00200CrossRefGoogle ScholarPubMed
Zuo, X. N., Ehmke, R., Mennes, M., Imperati, D., Castellanos, F. X., Sporns, O., & Milham, M. P. (2012). Network centrality in the human functional connectome. Cerebral Cortex, 22(8), 18621875. doi: 10.1093/cercor/bhr269CrossRefGoogle ScholarPubMed
Supplementary material: File

Wang et al. supplementary material

Wang et al. supplementary material

Download Wang et al. supplementary material(File)
File 2.9 MB