Zum Hauptinhalt springen

Exploring Host Genetic Polymorphisms Involved in SARS-CoV Infection Outcomes: Implications for Personalized Medicine in COVID-19.

Ramos-Lopez, O ; Daimiel, L ; et al.
In: International journal of genomics, Jg. 2020 (2020-10-19), S. 6901217
Online academicJournal

Exploring Host Genetic Polymorphisms Involved in SARS-CoV Infection Outcomes: Implications for Personalized Medicine in COVID-19  1. Background

Objective. To systematically explore genetic polymorphisms associated with the clinical outcomes in SARS-CoV infection in humans. Methods. This comprehensive literature search comprised available English papers published in PubMed/Medline and SCOPUS databases following the PRISMA-P guidelines and PICO/AXIS criteria. Results. Twenty-nine polymorphisms located in 21 genes were identified as associated with SARS-CoV susceptibility/resistance, disease severity, and clinical outcomes predominantly in Asian populations. Thus, genes implicated in key pathophysiological processes such as the mechanisms related to the entry of the virus into the cell and the antiviral immune/inflammatory responses were identified. Conclusions. Although caution must be taken, the results of this systematic review suggest that multiple genetic polymorphisms are associated with SARS-CoV infection features by affecting virus pathogenesis and host immune response, which could have important applications for the study and understanding of genetics in SARS-CoV-2/COVID-19 and for personalized translational clinical practice depending on the population studied and associated environments.

Coronaviruses (CoVs) have become one of the leading pathogens of the latest emerging outbreaks of respiratory disease, representing a serious public health burden worldwide [[1]]. A novel coronavirus was identified to play a crucial role in the severe acute respiratory syndrome (SARS) in 2003 [[2]]. Later, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which produces the disease coronavirus-2019 (COVID-19), has emerged in December 2019. This new virus appears to be highly contagious and has spread rapidly throughout the world, reaching a pandemic state, with important social and health system costs [[3]].

CoVs are a large family of positive-stranded, single-stranded, enveloped RNA viruses [[4]]. Relevant pathophysiological mechanisms of CoV, including SARS-CoV and SARS-CoV-2, are related to an exacerbated immunological response that the viral infection produces in the host [[5]]. In some cases, this reaction is excessive (inflammatory cytokine storm), triggering an extensive tissue damage and body dysfunction [[6]]. The clinical manifestations of this infection include typical symptoms such as cough, fever, asthenia, and mild respiratory distress. However, more serious respiratory injuries include pneumonia, acute respiratory distress syndrome, and respiratory failure, accompanied by inflammatory outcomes affecting adipokines and other mediators [[6]]. Furthermore, recent investigations have reported the presence of neurological, renal, hepatic, and cardiac complications in some patients [[7]]. On the other hand, a group of patients are asymptomatic or with very mild manifestations during the course of the infection, with a relatively short recovery time [[8]].

Certain risk factors for the chronicity and severity of COVID-19 infection have been reported, including age (over 60 years), male gender, and the presence of concomitant metabolic conditions such as obesity, diabetes, and hypertension [[9]]. Nevertheless, there is a constant search for biological factors associated with the evolution of COVID-19. Evidence suggests that genetic factors may influence the onset and progression of infectious diseases [[11]]. In this context, a number of genetic variants, mainly single nucleotide polymorphisms (SNPs), have been associated with the susceptibility/resistance to viral respiratory infections [[12]]. Until now, the specific role of the genetic make-up in SARS-CoV-2/COVID-19 disease has been insufficiently analyzed, although proinflammatory-related pathways have been involved [[6]].

Understanding the host genetic factors involved in SARS-CoV infection could contribute to the identification of new therapeutic tools for advancing in the prevention and clinical management of this disease through a personalized medicine approach [[13]]. Due to the lack of information concerning genetics and SARS-CoV-2/COVID-19 infection, the aim of the present review was to systematically explore the genetic polymorphisms associated with the clinical outcomes in SARS-CoV infection in humans in order to translate this information to SARS-CoV-2/COVID-19 research and clinical implementation.

2. Methods

A systematic review was performed in March 2020 to analyze the association of genetic polymorphisms with SARS-CoV infection outcomes. The methodological procedures of this systematic review were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols (PRISMA-P) guidelines [[14]]. Observational (cross-sectional, cohort, or case-control) studies exploring the role of host genetic polymorphisms in SARS-CoV disease (infection susceptibility, disease progression, and clinical outcomes) in adult subjects were included. Articles written only in the English language were selected. Duplicates, reviews, clinical trials, in vitro assays, animal experiments, and human studies reporting negative results or focused on other infectious diseases or genetic factors were excluded.

The comprehensive literature search encompassed available papers compiled in PubMed/Medline and SCOPUS databases without a period of time or study population restrictions. Advanced search techniques for each database were used to make the search more efficient (i.e., "MeSH" and "tiab" terms). The keywords used were as follows: ("genetic polymorphisms") OR ("genetic variations") OR ("gene polymorphisms") OR ("genetic variations") AND ("SARS-CoV") OR ("SARS CoV") ("SARS coronavirus") OR ("SARS virus"). The reference sections of the included articles were also scrutinized. This search strategy was replicated at different times to guarantee reproducibility. Moreover, two researchers performed independently the research. Using these terms, a total of 202 articles were identified. Five additional articles were added from reviews, which did not appear in this first stage (Figure 1). First, duplicates were eliminated ( n=132 ), obtaining 75 records for the eligibility criteria assessment. After the duplicates were removed, the titles, abstracts, and keywords of 75 manuscripts were screened, excluding those that did not meet the eligibility criteria ( n=41 ) or reported negative/inconsistent findings ( n=8 ). Finally, 26 articles were included in the current analyses (Figure 1).

Graph: Figure 1 PRISMA flow diagram summarizing the selection of papers included in this systematic review.

The most relevant information about each of the 26 selected studies included in the present review was analyzed using the Population, Intervention, Comparison, Outcome (PICO) criteria [[15]]. The selected articles met at least 3 inclusion criteria (Supplementary Table 1). The quality assessment of the analyzed studies was performed using the Appraisal tool for Cross-Sectional Studies (AXIS tool), a validated 20-point questionnaire with 6 subdomains (introduction, methods, results, discussion, and others) addressing study quality and reporting encompassing study design, sample size justification, target population, sampling frame, sample selection, measurement of validity/reliability, and overall methods [[16]]. Because the AXIS tool does not provide a numerical scale for assessing the quality of the study, subjective assessments by the authors are required [[16]]. Thus, the authors of this study performed a consensus procedure to evaluate the general and overall quality of records in order to select those to be included in the systematic review. The screening was completed with references coming from citations of the 26 selected studies. The results of the AXIS tool are summarized (Supplementary Table 2).

3. Results

The present systematic review revealed that 29 polymorphisms located in 21 genes were associated with SARS-CoV susceptibility/resistance, disease severity, and clinical manifestations/outcomes (Table 1).

Table 1 List of relevant polymorphisms in genes implicated in the susceptibility and progression to SARS-CoV infection.

GeneSNPRisk/protective alleleOutcomeFrequencyPopulationReference
Mechanisms of entry of the virus into the cell
ACE1Insertion/deletion (I/D)DHigher frequency of hypoxemia0.33Vietnamese[18]
CD209-336A>GGLower blood levels of lactate-dehydrogenase (an independent prognostic indicator for severity of SARS-CoV infection)0.10Chinese[47-48]
CLEC4MTandem repeat domain in exon 4HomozygotesLower susceptibility to SARS-CoV infection incidence0.46Chinese[17]
Immune response/inflammation phenomena
ICAM-3Asp143GlyGlyHigher lactate dehydrogenase levels and lower white blood cell counts0.10-0.17Chinese[49-50]
IFN-γ+874 A/TAHigher susceptibility to SARS-CoV infection incidence0.83Chinese[19-20]
RANTES-28C/GGHigher susceptibility to SARS-CoV infection incidence and increased risk of death0.21Chinese[20, 51]
OAS-1A/G SNP in exon 3GHigher susceptibility to SARS-CoV infection incidence0.48Vietnamese[22]
OAS-1A/G SNP at the 3'UTR 347 locus of the exon 8GLower susceptibility to SARS-CoV infection incidence0.24Chinese[52]
MxA-123C>AALower susceptibility to SARS-CoV infection incidence0.11Chinese[21]
MxAG/T SNP at position −88 in the promoter regionGHigher frequency of hypoxemia0.76-0.82Chinese, Vietnamese[21-22]
MxAG/T SNP at position −88 in the promoter regionGT genotypeHigher susceptibility to SARS-CoV infection incidence0.73-0.81Chinese[52-53]
IL-12 RB1+1664 C/TTHigher susceptibility to SARS-CoV infection incidence0.29Chinese[54]
FcgRIIaH131RRAssociated with a severe course of the disease (requiring treatment in an intensive care unit)0.34Chinese[55]
MLBCodon 54 variant (A/B)BHigher susceptibility to SARS-CoV infection incidence0.34-0.35Chinese[56-57]
CCL2G-2518AGHigher susceptibility to SARS-CoV infection incidence0.80Chinese[57]
AHSG−799A/TTHigher susceptibility to SARS-CoV infection incidence0.21Chinese[58]
TNF-α-204CT genotypeLower susceptibility to SARS-CoV infection incidence and increased risk of femoral head necrosis0.05Chinese[23]
TNF-α-308G/AAIncreased risk of femoral head necrosis0.06Chinese[23]
CD14-159C/TCAssociated with a severe course of the disease (requiring treatment in an intensive care unit)0.31Chinese[24]
HLA-BB

0703
B

0703
Higher susceptibility to SARS-CoV infection incidence0.11Chinese[25]
HLA-DRB1DRB1

0301
DRB1

0301
Lower susceptibility to SARS-CoV infection incidence0.01Chinese[25]
HLA-BB

4601
B

4601
Associated with severe cases of SARS-CoV infection (deceased or intubated patients)0.60Taiwanese[26]
HLA-DRB4DRB4

01010101
DRB4

01010101
Higher susceptibility to SARS-CoV infection incidence0.51Chinese[59]
HLA-BB

1502
B

1502
Lower susceptibility to SARS-CoV infection incidence0.26Chinese[59]
HLA-DRB3DRB3

030101
DRB3

030101
Lower susceptibility to SARS-CoV infection incidence0.28Chinese[59]
HLA-DRB1DRB1

1202
DRB1

1202
Higher susceptibility to SARS-CoV infection incidence0.47Vietnamese[60]
HLA-CwCw

0801
Cw

0801
Higher susceptibility to SARS-CoV infection incidence0.23Taiwanese[27]
HLA-CwCw

1502
Cw

1502
Lower susceptibility to SARS-CoV infection incidence0.10Taiwanese[28]
HLA-DRDR

0301
DR

0301
Lower susceptibility to SARS-CoV infection incidence0.11Taiwanese[28]

Available studies used mainly a gene-candidate approach to select those genes involved in disease pathogenesis and phenotypes based on a priori knowledge. Thus, genes implicated in key physiological processes such as the mechanisms related to the entry of the virus into the cell and the antiviral immune/inflammatory response were found (Table 1).

Of note, research was predominantly performed in Asian populations, including Chinese, Vietnamese, and Taiwanese populations. Moreover, genetic polymorphisms were mainly SNPs in coding and promoter regions, although insertion/deletion and tandem repeats were also described.

For instance, Chinese individuals homozygous for the tandem repeat domain in exon 4 of the CLEC4M gene were less susceptible to SARS infection [[17]]. Also, the insertion/deletion (I/D) polymorphism in the ACE1 gene was associated with the progression of pneumonia in SARS patients [[18]]. Both genes are involved in virus entry.

The rest of the genetic polymorphisms were located in cytokine and chemokine genes implicated in the immune response and inflammatory processes. For example, the IFN-γ +874A allele was associated with the susceptibility to develop SARS in two independent populations [[19]]. Also, the minor alleles of the -88G>T and -123C>A MxA promoter SNPs were significantly associated with a lower risk of SARS-CoV infection in Chinese [[21]]. Nevertheless, the G/T SNP at position −88 in the promoter region of this gene was associated with SARS-CoV disease progression [[22]].

Genetic variations in the inflammatory TNF-α gene were also screened. Thus, whereas the CT genotype at the TNF-α -204 locus of this gene was found to be associated with a protective effect on SARS, a higher A allele frequency of the TNF-α -308G/A polymorphism was found in the SARS population when compared with healthy controls [[23]]. Moreover, the prevalence of the CD14-159CC polymorphism was significantly higher in patients with severe SARS than in those with mild stages of the disease or controls [[24]].

The distribution of HLA alleles has been widely used as a strategy in the search for the etiology of infectious diseases. In this sense, three genetic polymorphisms in the HLA-B gene were related to different SARS-CoV outcomes, including infection predisposition, a protective phenotype [[25]], and severe forms of the disease [[26]]. In addition, the Cw 0801 and Cw 1502 variants in the HLA-Cw gene were associated with SARS risk [[27]] and SARS resistance [[28]] in Taiwanese, respectively.

4. Discussion

Personalized medicine involves a balanced knowledge of genotype, phenotype, and clinical features of the patient [[13]], where susceptibility to exacerbated infection is often dependent. Similar to other viral diseases, the first step in SARS-CoV infection includes the attachment of the virus to the host cell receptors [[29]]. In the present review, genetic variants in ACE1, CLEC4M, and CD209 genes were associated with SARS-CoV-related disease pathogenesis. Particularly, the angiotensin-converting enzyme 2 (ACE2, a homologue of ACE1) has been identified as a functional receptor for SARS-CoV [[30]]. Also, the C-type lectin domain family 4 member M (CLEC4M, also known as L-SIGN) was recognized as a binding receptor for this virus [[31]]. CD209 (also known as DC-SIGN) shares amino acid identity with CLEC4M, which was involved in facilitating SARS-CoV viral transmission to susceptible cells [[32]]. These findings demonstrate the important functions of these molecules in the virus entry. Nevertheless, two additional studies failed to demonstrate a relationship between ACE2 polymorphisms and SARS-CoV outcome in Asians so far [[33]], emphasizing the need for future research in other populations.

Virus survival and virus-induced disease outcomes (including SARS-CoV) are commonly linked to the modulation of the immune response in the host [[35]]. As expected, in this review, genetic polymorphisms in genes involved in immunocompetence/inflammatory pathways were found (ICAM-3, IFN-γ, RANTES, OAS-1, MxA, IL-12 RB1, FcgRIIa, MLB, CCL2, AHSG, TNF-α, CD14). Most of them are cytokines or chemokines that are known to be important mediators of early defense against infections, activating protective mechanisms in infected cells [[36]]. These processes occur during the immune response driven by the SARS-CoV infection and comprise T cell stimulation (ICAM-3, IL-12 RB1), macrophage activation/deactivation (IFN-γ, AHSG), cell trafficking (RANTES, CCL2), RNA degradation and the inhibition of viral replication (OAS-1, MxA), link between humoral and cell-mediated immune reaction (FcgRIIa), virus binding (MLB), and inflammation (TNF-α, CD14) (http://www.genecards.org).

Genetic variations in HLA class I or II genes have been often associated with susceptibility/resistance to a wide range of infections, including SARS-CoV [[37]]. Accordingly, the genes of the HLA complex were associated with SARS-CoV outcomes in this study (HLA-B, HLA-DRB1, HLA-DRB4, HLA-DRB3, HLA-Cw). HLA play a critical role in the presentation of antigenic peptides to T cells, orchestrating an immune response aimed at removing nonself material via neutralization of antibodies, cytokines, and activated cytotoxic T cells [[38]].

Robust evidence support that genetic variation in human populations contributes to the onset and development of several chronic diseases, including those of an infectious nature [[39]]. The results of this systematic review show that research exploring the genetic contribution to SARS-CoV infection was predominantly performed in Asian populations, which may be related to its epidemiological origin in China in 2002 [[40]]. However, allele and genotype frequencies of genetic variants may vary depending on region; therefore, the results should not be extrapolated to other populations without prior exploration. Therefore, further investigation is required to determine the pattern of distribution of these and other candidate polymorphisms in other groups as well as confirm associations with SARS-CoV outcomes. This knowledge is particularly important in populations with heterogenic heritages exposed to absolutely different environmental factors. An example could be Latin American countries, including Mexico, which have an admixture genome with Amerindian, European, and African ancestries; a high prevalence of obesity; and associated comorbidities as well as the adoption of an unhealthy lifestyle (high-fat/sugar diet and physical inactivity) that could exacerbate the outcome of viral infections [[41]]. Or even some SNPs that have not been associated with SARS-CoV disease outcomes in the Asian population could become a significant association in other populations because of a strong environmental pressure. Thus, we cannot rule out the possibility that other SNPs in other genes could play a role in susceptibility to SARS-CoV infection in these populations.

In this context, personalized medicine is an integrative therapeutic approach that considers conventional factors (age, gender, clinical phenotype), as well as emerging genetics and interactions with environmental factors to individualize prevention, diagnosis, treatments, and prognosis [[42]]. Applying the principles of personalized medicine in the current care schemes for the control of infectious diseases (i.e., SARS-CoV) could allow the identification of genetically susceptible groups, disease risk prediction, and personalized therapies to reduce infection-related complications, high mortality rates, and the optimization of economic resources for health care and individualized management of inflammation (Figure 2).

Graph: Figure 2 Genetic background and other precision parameters as important predictors of SARS-CoV infection and personalized translational medicine interventions.

Currently, the high incidence of SARS-CoV-2/COVID-19 around the world, the speed of its spread, and the absence of a specific drug for its pharmacological management emphasize the need to identify risk factors associated with the dynamics of infection and disease progression in order to mitigate its negative impact on the society. A better understanding of the relationship between the genetic make-up and SARS-CoV-2/COVID-19 will provide new insights into the disease pathogenesis by explaining particular phenotypes and clinical responses. Moreover, this knowledge will also aid in identifying biomarkers as potential therapeutic targets for evaluating the efficacy of genome-based interventions and other personalized treatments within the new era of precision medicine.

In addition to the polymorphisms analyzed in this review, new genetic variants affecting SARS-CoV and SARS-CoV-2/COVID-19 susceptibility need to be further explored as well as the interplay of other emerging parameters of individualization such as epigenetic signatures, metabolomic fingerprints, metagenomic data, and lifestyle factors through an integrative holistic approach [[43]]. Potential gene×drug or gene×inflammation interactions also deserve further investigation. In this regard, drugs targeting the inflammatory cytokines IL-1 and IL-6 have been proposed to reduce extreme immune reaction to the virus and extensive tissue damage [[44]], where genetics may play a pivotal role. Additionally, genotyping of virus isolates to detect specific multiple mutations is of great importance for the understanding of the evolution and transmission of SARS-CoV infections as well as for vaccine development and disease control [[46]].

5. Conclusion

Although caution must be taken, the results of this systematic review suggest that multiple genetic polymorphisms are associated with SARS-CoV infection outcomes (susceptibility and severity, hospitalization, ICU stays, and medications) by affecting virus pathogenesis and host immune response, which could have important applications for the study and understanding of genetics in SARS-CoV-2/COVID-19 and for personalized translational clinical practice depending on the population studied and associated environments.

Data Availability

All data are available in the article.

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this paper.

Authors' Contributions

ORL and JAM performed the search of articles and wrote the draft of the manuscript. LD, ARM, DMU, and JAV contributed to the analysis and critical interpretation of the data. All authors read and approved the final manuscript.

Acknowledgments

The support from CIBERObn, IMDEA-Food Institute, and HM hospitals is gratefully acknowledged.

Supplementary Materials

Supplementary Table 1. PICO criteria used for the inclusion and exclusion of screened studies. Supplementary Table 2. Quality assessment of the individual studies by applying the AXIS tool.

REFERENCES 1 de Wit E., van Doremalen N., Falzarano D., Munster V. J. SARS and MERS: recent insights into emerging coronaviruses. Nature Reviews Microbiology. 2016; 14(8): 523-534, 10.1038/nrmicro.2016.81, 2-s2.0-84976271703, 27344959 2 Ksiazek T. G., Erdman D., Goldsmith C. S., Zaki S. R., Peret T., Emery S., Tong S., Urbani C., Comer J. A., Lim W., Rollin P. E., Dowell S. F., Ling A. E., Humphrey C. D., Shieh W. J., Guarner J., Paddock C. D., Rota P., Fields B., DeRisi J., Yang J. Y., Cox N., Hughes J. M., LeDuc J., Bellini W. J., Anderson L. J., SARS Working Group. A novel coronavirus associated with severe acute respiratory syndrome. The New England Journal of Medicine. 2003; 348(20): 1953-1966, 10.1056/NEJMoa030781, 2-s2.0-0038076030, 12690092 3 Hamid S., Mir M. Y., Rohela G. K. Novel coronavirus disease (COVID-19): a pandemic (epidemiology, pathogenesis and potential therapeutics). New Microbes and New Infections. 2020; 35, article 100679, 10.1016/j.nmni.2020.100679 4 Madhugiri R., Fricke M., Marz M., Ziebuhr J. Coronavirus cis-acting RNA elements. Advances in Virus Research. 2016; 96, 127-163, 10.1016/bs.aivir.2016.08.007, 2-s2.0-84995532215 5 Merad M., Martin J. C. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nature Reviews Immunology. 2020; 20(6): 355-362, 10.1038/s41577-020-0331-4, 32376901 6 Ye Q., Wang B., Mao J. The pathogenesis and treatment of the 'Cytokine Storm' in COVID-19. The Journal of Infection. 2020; 80(6): 607-613, 10.1016/j.jinf.2020.03.037, 32283152 7 Atri D., Siddiqi H. K., Lang J. P., Nauffal V., Morrow D. A., Bohula E. A. COVID-19 for the cardiologist: basic virology, epidemiology, cardiac manifestations, and potential therapeutic strategies. JACC: Basic to Translational Science. 2020; 5(5): 518-536, 10.1016/j.jacbts.2020.04.002, 32292848 8 Chen Y., Liu Q., Guo D. Emerging coronaviruses: genome structure, replication, and pathogenesis. Journal of Medical Virology. 2020; 92(4): 418-423, 10.1002/jmv.25681, 31967327 9 Watanabe M., Risi R., Tuccinardi D., Baquero C. J., Manfrini S., Gnessi L. Obesity and SARS‐CoV‐2: a population to safeguard. Diabetes/Metabolism Research and Reviews. 2020; 36(7, article e3325), 10.1002/dmrr.3325 Conti P., Younes A. Coronavirus COV-19/SARS-CoV-2 affects women less than men: clinical response to viral infection. Journal of Biological Regulators and Homeostatic Agents. 2020; 34(2): 339-343, 10.23812/Editorial-Conti-3, 32253888 Hill A. V. S. Evolution, revolution and heresy in the genetics of infectious disease susceptibility. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 2012; 367(1590): 840-849, 10.1098/rstb.2011.0275, 2-s2.0-84856654988, 22312051 Patarčić I., Gelemanović A., Kirin M., Kolčić I., Theodoratou E., Baillie K. J., de Jong M. D., Rudan I., Campbell H., Polašek O. The role of host genetic factors in respiratory tract infectious diseases: systematic review, meta-analyses and field synopsis. Scientific Reports. 2015; 5(1), 10.1038/srep16119, 2-s2.0-84946239477 Ramos-Lopez O., Milagro F. I., Allayee H., Chmurzynska A., Choi M. S., Curi R., de Caterina R., Ferguson L. R., Goni L., Kang J. X., Kohlmeier M., Marti A., Moreno L. A., Pérusse L., Prasad C., Qi L., Reifen R., Riezu-Boj J. I., San-Cristobal R., Santos J. L., Martínez J. A. Guide for current nutrigenetic, nutrigenomic, and nutriepigenetic approaches for precision nutrition involving the prevention and management of chronic diseases associated with obesity. Journal of Nutrigenetics and Nutrigenomics. 2017; 10(1-2): 43-62, 10.1159/000477729, 2-s2.0-85023776188 Liberati A., Altman D. G., Tetzlaff J., Mulrow C., Gøtzsche P. C., Ioannidis J. P. A., Clarke M., Devereaux P. J., Kleijnen J., Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. Journal of Clinical Epidemiology. 2009; 62(10): e1-34, 10.1016/j.jclinepi.2009.06.006, 2-s2.0-84872011968, 19631507 Boudin F., Nie J. Y., Bartlett J. C., Grad R., Pluye P., Dawes M. Combining classifiers for robust PICO element detection. BMC Medical Informatics and Decision Making. 2010; 10(1): 29, 10.1186/1472-6947-10-29, 2-s2.0-77952104897 Downes M. J., Brennan M. L., Williams H. C., Dean R. S. Development of a critical appraisal tool to assess the quality of cross-sectional studies (AXIS). BMJ Open. 2016; 6(12, article e011458), 10.1136/bmjopen-2016-011458, 2-s2.0-85004045202, 27932337 Chan V. S. F., Chan K. Y. K., Chen Y., Poon L. L. M., Cheung A. N. Y., Zheng B., Chan K. H., Mak W., Ngan H. Y. S., Xu X., Screaton G., Tam P. K. H., Austyn J. M., Chan L. C., Yip S. P., Peiris M., Khoo U. S., Lin C. L. S. Homozygous L-SIGN (CLEC4M) plays a protective role in SARS coronavirus infection. Nature Genetics. 2006; 38(1): 38-46, 10.1038/ng1698, 2-s2.0-29444444113, 16369534 Itoyama S., Keicho N., Quy T., Phi N. C., Long H. T., Ha L. D., Ban V. V., Ohashi J., Hijikata M., Matsushita I., Kawana A., Yanai H., Kirikae T., Kuratsuji T., Sasazuki T. ACE1 polymorphism and progression of SARS. Biochemical and Biophysical Research Communications. 2004; 323(3): 1124-1129, 10.1016/j.bbrc.2004.08.208, 2-s2.0-4544366685, 15381116 Chong W. P., Ip W. K. E., Tso G. H. W., Ng M. W., Wong W. H. S., Law H. K. W., Yung R. W. H., Chow E. Y., Au K. L., Chan E. Y. T., Lim W., Peiris J. S. M., Lau Y. L. The interferon gamma gene polymorphism +874 A/T is associated with severe acute respiratory syndrome. BMC Infectious Diseases. 2006; 6(1): 82, 10.1186/1471-2334-6-82, 2-s2.0-33646891057 Lau Y. L., Peiris J. S. Association of cytokine and chemokine gene polymorphisms with severe acute respiratory syndrome. Hong Kong Medical Journal. 2009; 15Supplement 2, 43-46 Ching J. C. Y., Chan K. Y. K., Lee E. H. L., Xu M. S., Ting C. K. P., So T. M. K., Sham P. C., Leung G. M., Peiris J. S. M., Khoo U. S. Significance of the myxovirus resistance A (MxA) gene -123C>a single-nucleotide polymorphism in suppressed interferon β induction of severe acute respiratory syndrome coronavirus infection. The Journal of Infectious Diseases. 2010; 201(12): 1899-1908, 10.1086/652799, 2-s2.0-77952560927, 20462354 Hamano E., Hijikata M., Itoyama S., Quy T., Phi N. C., Long H. T., Ha L. D., Ban V. V., Matsushita I., Yanai H., Kirikae F., Kirikae T., Kuratsuji T., Sasazuki T., Keicho N. Polymorphisms of interferon-inducible genes OAS-1 and MxA associated with SARS in the Vietnamese population. Biochemical and Biophysical Research Communications. 2005; 329(4): 1234-1239, 10.1016/j.bbrc.2005.02.101, 2-s2.0-20244368345, 15766558 Wang S., Wei M., Han Y., Zhang K., He L., Yang Z., Su B., Zhang Z., Hu Y., Hui W. Roles of TNF-α gene polymorphisms in the occurrence and progress of SARS-Cov infection: a case-control study. BMC Infectious Diseases. 2008; 8(1): 27, 10.1186/1471-2334-8-27, 2-s2.0-44049086127 Yuan F. F., Boehm I., Chan P. K. S., Marks K., Tang J. W., Hui D. S. C., Sung J. J. Y., Dyer W. B., Geczy A. F., Sullivan J. S. High prevalence of the CD14-159CC genotype in patients infected with severe acute respiratory syndrome-associated coronavirus. Clinical and Vaccine Immunology. 2007; 14(12): 1644-1645, 10.1128/CVI.00100-07, 2-s2.0-38049004890, 17913858 Ng M. H. L., Lau K. M., Li L., Cheng S. H., Chan W. Y., Hui P. K., Zee B., Leung C. B., Sung J. J. Y. Association of human-leukocyte-antigen class I (B0703) and class II (DRB10301) genotypes with susceptibility and resistance to the development of severe acute respiratory syndrome. The Journal of Infectious Diseases. 2004; 190(3): 515-518, 10.1086/421523, 2-s2.0-3242774516, 15243926 Lin M., Tseng H. K., Trejaut J. A., Lee H. L., Loo J. H., Chu C. C., Chen P. J., Su Y. W., Lim K. H., Tsai Z. U., Lin R. Y., Lin R. S., Huang C. H. Association of HLA class I with severe acute respiratory syndrome coronavirus infection. BMC Medical Genetics. 2003; 4(1): 9, 10.1186/1471-2350-4-9, 2-s2.0-2542493298 Chen Y. M. A., Liang S. Y., Shih Y. P., Chen C. Y., Lee Y. M., Chang L., Jung S. Y., Ho M. S., Liang K. Y., Chen H. Y., Chan Y. J., Chu D. C. Epidemiological and genetic correlates of severe acute respiratory syndrome coronavirus infection in the hospital with the highest nosocomial infection rate in Taiwan in 2003. Journal of Clinical Microbiology. 2006; 44(2): 359-365, 10.1128/JCM.44.2.359-365.2006, 2-s2.0-32344449457, 16455884 Wang S. F., Chen K. H., Chen M., Li W. Y., Chen Y. J., Tsao C. H., Yen M. Y., Huang J. C., Chen Y. M. A. Human-leukocyte antigen class I Cw 1502 and class II DR 0301 genotypes are associated with resistance to severe acute respiratory syndrome (SARS) infection. Viral Immunology. 2011; 24(5): 421-426, 10.1089/vim.2011.0024, 2-s2.0-80054733924, 21958371 Hofmann H., Pöhlmann S. Cellular entry of the SARS coronavirus. Trends in Microbiology. 2004; 12(10): 466-472, 10.1016/j.tim.2004.08.008, 2-s2.0-4644323974, 15381196 Li W., Moore M. J., Vasilieva N., Sui J., Wong S. K., Berne M. A., Somasundaran M., Sullivan J. L., Luzuriaga K., Greenough T. C., Choe H., Farzan M. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003; 426(6965): 450-454, 10.1038/nature02145, 2-s2.0-0344395657, 14647384 Jeffers S. A., Tusell S. M., Gillim-Ross L., Hemmila E. M., Achenbach J. E., Babcock G. J., Thomas W. D., Thackray L. B., Young M. D., Mason R. J., Ambrosino D. M., Wentworth D. E., DeMartini J. C., Holmes K. V. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus. Proceedings of the National Academy of Sciences of the United States of America. 2004; 101(44): 15748-15753, 10.1073/pnas.0403812101, 2-s2.0-8144221600, 15496474 Yang Z. Y., Huang Y., Ganesh L., Leung K., Kong W. P., Schwartz O., Subbarao K., Nabel G. J. pH-dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC-SIGN. Journal of Virology. 2004; 78(11): 5642-5650, 10.1128/JVI.78.11.5642-5650.2004, 2-s2.0-2442691605, 15140961 Chiu R. W. K., Tang N. L. S., Hui D. S. C., Chung G. T. Y., Chim S. S. C., Chan K. C. A., Sung Y. M., Chan L. Y. S., Tong Y. K., Lee W. S., Chan P. K. S., Lo Y. M. D. ACE2 gene polymorphisms do not affect outcome of severe acute respiratory syndrome. Clinical Chemistry. 2004; 50(9): 1683-1686, 10.1373/clinchem.2004.035436, 2-s2.0-4344682612, 15331509 Itoyama S., Keicho N., Hijikata M., Quy T., Phi N. C., Long H. T., Ha L. D., Ban V. V., Matsushita I., Yanai H., Kirikae F., Kirikae T., Kuratsuji T., Sasazuki T. Identification of an alternative 5'-untranslated exon and new polymorphisms of angiotensin-converting enzyme 2 gene: lack of association with SARS in the Vietnamese population. American Journal of Medical Genetics. Part A. 2005; 136(1): 52-57, 10.1002/ajmg.a.30779, 2-s2.0-21644480682, 15937940 Braciale T. J., Hahn Y. S. Immunity to viruses. Immunological Reviews. 2013; 255(1): 5-12, 10.1111/imr.12109, 2-s2.0-84881648617, 23947343 Biron C. A. Role of early cytokines, including alpha and beta interferons (IFN-alpha/beta), in innate and adaptive immune responses to viral infections. Seminars in Immunology. 1998; 10(5): 383-390, 10.1006/smim.1998.0138, 2-s2.0-0032191176, 9799713 Martin M. P., Carrington M. Immunogenetics of viral infections. Current Opinion in Immunology. 2005; 17(5): 510-516, 10.1016/j.coi.2005.07.012, 2-s2.0-23944474386, 16084708 Blackwell J. M., Jamieson S. E., Burgner D. HLA and infectious diseases. Clinical Microbiology Reviews. 2009; 22(2): 370-385, 10.1128/CMR.00048-08, 2-s2.0-66449133984, 19366919 Burgner D., Jamieson S. E., Blackwell J. M. Genetic susceptibility to infectious diseases: big is beautiful, but will bigger be even better?. The Lancet Infectious Diseases. 2006; 6(10): 653-663, 10.1016/S1473-3099(06)70601-6, 2-s2.0-33748902565, 17008174 Xu R. H., He J. F., Evans M. R., Peng G. W., Field H. E., Yu D. W., Lee C. K., Luo H. M., Lin W. S., Lin P., Li L. H., Liang W. J., Lin J. Y., Schnur A. Epidemiologic clues to SARS origin in China. Emerging Infectious Diseases. 2004; 10(6): 1030-1037, 10.3201/eid1006.030852, 2-s2.0-2542611561, 15207054 Ramos-Lopez O., Martinez-Lopez E., Roman S., Fierro N. A., Panduro A. Genetic, metabolic and environmental factors involved in the development of liver cirrhosis in Mexico. World Journal of Gastroenterology. 2015; 21(41): 11552-11566, 10.3748/wjg.v21.i41.11552, 2-s2.0-84947262445, 26556986 Offit K. Personalized medicine: new genomics, old lessons. Human Genetics. 2011; 130(1): 3-14, 10.1007/s00439-011-1028-3, 2-s2.0-79959990099, 21706342 Goni L., Cuervo M., Milagro F. I., Martínez J. A. Future perspectives of personalized weight loss interventions based on nutrigenetic, epigenetic, and metagenomic data. The Journal of Nutrition. 2015; 146(4): 905S-912S, 10.3945/jn.115.218354, 2-s2.0-84963815239 Conti P., Gallenga C. E., Tetè G., Al Caraffa G. R., Younes A., Toniato E., Ross R., Kritas S. K. How to reduce the likelihood of coronavirus-19 (CoV-19 or SARS-CoV-2) infection and lung inflammation mediated by IL-1. Journal of Biological Regulators and Homeostatic Agents. 2020; 34, 32228825, 10.23812/Editorial-Conti-2 Buonaguro F. M., Puzanov I., Ascierto P. A. Anti-IL6R role in treatment of COVID-19-related ARDS. Journal of Translational Medicine. 2020; 18(1): 165, 10.1186/s12967-020-02333-9, 32290847 Yin C. Genotyping coronavirus SARS-CoV-2: methods and implications. Genomics. 2020; 112(5): 3588-3596, 10.1016/j.ygeno.2020.04.016, 32353474 Chan K. Y. K., Xu M. S., Ching J. C. Y., So T. M. K., Lai S. T., Chu C. M., Yam L. Y. C., Wong A. T. Y., Chung P. H., Chan V. S. F., Lin C. L. S., Sham P. C., Leung G. M., Peiris J. S. M., Khoo U. S. CD209 (DC-SIGN) -336A>G promoter polymorphism and severe acute respiratory syndrome in Hong Kong Chinese. Human Immunology. 2010; 71(7): 702-707, 10.1016/j.humimm.2010.03.006, 2-s2.0-77953917346, 20359516 Chan K. Y., Xu M. S., Ching J. C., Chan V. S., Ip Y. C., Yam L., Chu C. M., Lai S. T., So K. M., Wong T. Y., Chung P. H., Tam P., Yip S. P., Sham P., Lin C. L., Leung G. M., Peiris J. S., Khoo U. S. Association of a single nucleotide polymorphism in the CD209 (DC-SIGN) promoter with SARS severity. Hong Kong Medical Journal. 2010; 165 Supplement 4, 37-42, 20864747 Khoo U. S., Chan K. Y., Ching J. C. Y., Chan V. S., Ip Y. C., Yam L., Chu C. M., Lai S. T., So K. M., Wong T. Y., Chung P. H., Tam P., Yip S. P., Sham P., Leung G. M., Lin C. L., Peiris J. S. M. Functional role of ICAM-3 polymorphism in genetic susceptibility to SARS infection. Hong Kong Medical Journal. 2009; 15Supplement 6, 26-29, 19801714 Chan K. Y. K., Ching J. C. Y., Xu M. S., Cheung A. N. Y., Yip S. P., Yam L. Y. C., Lai S. T., Chu C. M., Wong A. T. Y., Song Y. Q., Huang F. P., Liu W., Chung P. H., Leung G. M., Chow E. Y. D., Chan E. Y. T., Chan J. C. K., Ngan H., Tam P., Chan L. C., Sham P., Chan V. S. F., Peiris M., Lin S. C. L., Khoo U. S. Association of ICAM3 genetic variant with severe acute respiratory syndrome. The Journal of Infectious Diseases. 2007; 196(2): 271-280, 10.1086/518892, 2-s2.0-34447292078, 17570115 Ng M. W., Zhou G., Chong W. P., Lee L. W. Y., Law H. K. W., Zhang H., Wong W. H. S., Fok S. F. S., Zhai Y., Yung R. W. H., Chow E. Y., Au K. L., Chan E. Y. T., Lim W., Peiris J. S. M., He F., Lau Y. L. The association of RANTES polymorphism with severe acute respiratory syndrome in Hong Kong and Beijing Chinese. BMC Infectious Diseases. 2007; 7(1): 50, 10.1186/1471-2334-7-50, 2-s2.0-34347374296 He J., Feng D., de Vlas S. J., Wang H., Fontanet A., Zhang P., Plancoulaine S., Tang F., Zhan L., Yang H., Wang T., Richardus J. H., Habbema J. D. F., Cao W. Association of SARS susceptibility with single nucleic acid polymorphisms of OAS1 and MxA genes: a case-control study. BMC Infectious Diseases. 2006; 6(1): 106, 10.1186/1471-2334-6-106, 2-s2.0-33747380255 Wang H. W., He J., Zhang P. H., Tang F., Wang T. B., Luan Y. H., Lv H., Cao Z. Y., Wu X. M., Zhao Q. M., Liu W., Cao W. C. A case-control study on the mxA polymorphisms and susceptibility to severe acute respiratory syndromes. Zhonghua Liu Xing Bing Xue Za Zhi. 2005; 26(8): 574-577, 16390004 Tang F., Liu W., Zhang F., Xin Z. T., Wei M. T., Zhang P. H., Yang H., Ly H., Cao W. C. IL-12 RB1 genetic variants contribute to human susceptibility to severe acute respiratory syndrome infection among Chinese. PLoS One. 2008; 3(5, article e2183), 10.1371/journal.pone.0002183, 2-s2.0-48249126829, 18478121 Yuan F. F., Tanner J., Chan P. K. S., Biffin S., Dyer W. B., Geczy A. F., Tang J. W., Hui D. S. C., Sung J. J. Y., Sullivan J. S. Influence of FcgammaRIIA and MBL polymorphisms on severe acute respiratory syndrome. Tissue Antigens. 2005; 66(4): 291-296, 10.1111/j.1399-0039.2005.00476.x, 2-s2.0-26044438261, 16185324 Zhang H., Zhou G., Zhi L., Yang H., Zhai Y., Dong X., Zhang X., Gao X., Zhu Y., He F. Association between mannose-binding lectin gene polymorphisms and susceptibility to severe acute respiratory syndrome coronavirus infection. The Journal of Infectious Diseases. 2005; 192(8): 1355-1361, 10.1086/491479, 2-s2.0-25844469142, 16170752 Tu X., Chong W. P., Zhai Y., Zhang H., Zhang F., Wang S., Liu W., Wei M., Siu N. H. O., Yang H., Yang W., Cao W., Lau Y. L., He F., Zhou G. Functional polymorphisms of the CCL2 and MBL genes cumulatively increase susceptibility to severe acute respiratory syndrome coronavirus infection. The Journal of Infection. 2015; 71(1): 101-109, 10.1016/j.jinf.2015.03.006, 2-s2.0-84930542538, 25818534 Zhu X., Wang Y., Zhang H., Liu X., Chen T., Yang R., Shi Y., Cao W., Li P., Ma Q., Zhai Y., He F., Zhou G., Cao C. Genetic variation of the human α-2-Heremans-Schmid glycoprotein (AHSG) gene associated with the risk of SARS-CoV infection. PLoS One. 2011; 6(8, article e23730), 10.1371/journal.pone.0023730, 2-s2.0-80051718944, 21904596 Ng M. H., Cheng S. H., Lau K. M., Leung G. M., Khoo U. S., Zee B. C., Sung J. J. Immunogenetics in SARS: a case-control study. Hong Kong Medical Journal. 2010; 165 Supplement 4, 29-33, 20864745 Keicho N., Itoyama S., Kashiwase K., Phi N. C., Long H. T., Ha L. D., Ban V. V., Hoa B. K., Hang N. T. L., Hijikata M., Sakurada S., Satake M., Tokunaga K., Sasazuki T., Quy T. Association of human leukocyte antigen class II alleles with severe acute respiratory syndrome in the Vietnamese population. Human Immunology. 2009; 70(7): 527-531, 10.1016/j.humimm.2009.05.006, 2-s2.0-67449149672, 19445991

By Omar Ramos-Lopez; Lidia Daimiel; Ana Ramírez de Molina; Diego Martínez-Urbistondo; Juan A. Vargas and J. Alfredo Martínez

Reported by Author; Author; Author; Author; Author; Author

Titel:
Exploring Host Genetic Polymorphisms Involved in SARS-CoV Infection Outcomes: Implications for Personalized Medicine in COVID-19.
Autor/in / Beteiligte Person: Ramos-Lopez, O ; Daimiel, L ; Ramírez de Molina, A ; Martínez-Urbistondo, D ; Vargas, JA ; Martínez, JA
Link:
Zeitschrift: International journal of genomics, Jg. 2020 (2020-10-19), S. 6901217
Veröffentlichung: [New York, NY] : Hindawi Publishing Corporation, [2013]-, 2020
Medientyp: academicJournal
ISSN: 2314-4378 (electronic)
DOI: 10.1155/2020/6901217
Sonstiges:
  • Nachgewiesen in: MEDLINE
  • Sprachen: English
  • Publication Type: Journal Article; Review
  • Language: English
  • [Int J Genomics] 2020 Oct 19; Vol. 2020, pp. 6901217. <i>Date of Electronic Publication: </i>2020 Oct 19 (<i>Print Publication: </i>2020).
  • References: Hong Kong Med J. 2010;16(5 Suppl 4):37-42. (PMID: 20864747) ; Nature. 2003 Nov 27;426(6965):450-4. (PMID: 14647384) ; Proc Natl Acad Sci U S A. 2004 Nov 2;101(44):15748-53. (PMID: 15496474) ; J Biol Regul Homeost Agents. 2020 Mar 31;34(2):333-338. (PMID: 32228825) ; Hum Genet. 2011 Jul;130(1):3-14. (PMID: 21706342) ; J Infect. 2015 Jul;71(1):101-9. (PMID: 25818534) ; Hong Kong Med J. 2010;16(5 Suppl 4):29-33. (PMID: 20864745) ; J Virol. 2004 Jun;78(11):5642-50. (PMID: 15140961) ; PLoS One. 2008 May 14;3(5):e2183. (PMID: 18478121) ; Curr Opin Immunol. 2005 Oct;17(5):510-6. (PMID: 16084708) ; J Infect Dis. 2010 Jun 15;201(12):1899-908. (PMID: 20462354) ; N Engl J Med. 2003 May 15;348(20):1953-66. (PMID: 12690092) ; Hong Kong Med J. 2009 Oct;15 Suppl 6:26-9. (PMID: 19801714) ; Clin Microbiol Rev. 2009 Apr;22(2):370-85, Table of Contents. (PMID: 19366919) ; World J Gastroenterol. 2015 Nov 7;21(41):11552-66. (PMID: 26556986) ; Zhonghua Liu Xing Bing Xue Za Zhi. 2005 Aug;26(8):574-7. (PMID: 16390004) ; Semin Immunol. 1998 Oct;10(5):383-90. (PMID: 9799713) ; Immunol Rev. 2013 Sep;255(1):5-12. (PMID: 23947343) ; Clin Vaccine Immunol. 2007 Dec;14(12):1644-5. (PMID: 17913858) ; PLoS One. 2011;6(8):e23730. (PMID: 21904596) ; Tissue Antigens. 2005 Oct;66(4):291-6. (PMID: 16185324) ; New Microbes New Infect. 2020 Apr 14;35:100679. (PMID: 32322401) ; J Transl Med. 2020 Apr 14;18(1):165. (PMID: 32290847) ; Trends Microbiol. 2004 Oct;12(10):466-72. (PMID: 15381196) ; J Clin Microbiol. 2006 Feb;44(2):359-65. (PMID: 16455884) ; Hum Immunol. 2009 Jul;70(7):527-31. (PMID: 19445991) ; Am J Med Genet A. 2005 Jul 1;136(1):52-7. (PMID: 15937940) ; BMC Infect Dis. 2008 Feb 29;8:27. (PMID: 18312678) ; J Infect Dis. 2005 Oct 15;192(8):1355-61. (PMID: 16170752) ; J Infect Dis. 2004 Aug 1;190(3):515-8. (PMID: 15243926) ; Diabetes Metab Res Rev. 2020 Apr 21;:e3325. (PMID: 32314503) ; Nat Rev Immunol. 2020 Jun;20(6):355-362. (PMID: 32376901) ; Emerg Infect Dis. 2004 Jun;10(6):1030-7. (PMID: 15207054) ; Philos Trans R Soc Lond B Biol Sci. 2012 Mar 19;367(1590):840-9. (PMID: 22312051) ; BMC Med Inform Decis Mak. 2010 May 15;10:29. (PMID: 20470429) ; Viral Immunol. 2011 Oct;24(5):421-6. (PMID: 21958371) ; BMC Infect Dis. 2006 Jul 06;6:106. (PMID: 16824203) ; J Clin Epidemiol. 2009 Oct;62(10):e1-34. (PMID: 19631507) ; BMC Infect Dis. 2007 Jun 01;7:50. (PMID: 17540042) ; J Med Virol. 2020 Apr;92(4):418-423. (PMID: 31967327) ; Hong Kong Med J. 2009 Feb;15 Suppl 2:43-6. (PMID: 19258635) ; Nat Rev Microbiol. 2016 Aug;14(8):523-34. (PMID: 27344959) ; J Infect. 2020 Jun;80(6):607-613. (PMID: 32283152) ; Adv Virus Res. 2016;96:127-163. (PMID: 27712622) ; Nat Genet. 2006 Jan;38(1):38-46. (PMID: 16369534) ; BMC Infect Dis. 2006 May 04;6:82. (PMID: 16672072) ; Clin Chem. 2004 Sep;50(9):1683-6. (PMID: 15331509) ; Sci Rep. 2015 Nov 03;5:16119. (PMID: 26524966) ; BMJ Open. 2016 Dec 8;6(12):e011458. (PMID: 27932337) ; Lancet Infect Dis. 2006 Oct;6(10):653-63. (PMID: 17008174) ; Biochem Biophys Res Commun. 2004 Oct 22;323(3):1124-9. (PMID: 15381116) ; JACC Basic Transl Sci. 2020 Apr 10;5(5):518-536. (PMID: 32292848) ; J Infect Dis. 2007 Jul 15;196(2):271-80. (PMID: 17570115) ; J Nutr. 2015 Apr 1;146(4):905S-912S. (PMID: 26962191) ; J Biol Regul Homeost Agents. 2020 Apr 07;34(2):339-343. (PMID: 32253888) ; Biochem Biophys Res Commun. 2005 Apr 22;329(4):1234-9. (PMID: 15766558) ; J Nutrigenet Nutrigenomics. 2017;10(1-2):43-62. (PMID: 28689206) ; Hum Immunol. 2010 Jul;71(7):702-7. (PMID: 20359516) ; Genomics. 2020 Sep;112(5):3588-3596. (PMID: 32353474) ; BMC Med Genet. 2003 Sep 12;4:9. (PMID: 12969506)
  • Entry Date(s): Date Created: 20201028 Latest Revision: 20220417
  • Update Code: 20240513
  • PubMed Central ID: PMC7582067

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -