Αρχειοθήκη ιστολογίου

Τετάρτη 28 Οκτωβρίου 2020

Association Between Human Papilloma Virus Infection and Malignant Sinonasal Inverted Papilloma

xloma.fota13 shared this article with you from Inoreader

Objective

This systematic review and meta‐analysis aimed to evaluate the risk of malignant sinonasal inverted papilloma (SNIP) according to the type of human papilloma virus (HPV) infection.

Methods

The databases of PubMed, EmBase, and Web of Science were searched for studies that reported the risk of malignant SNIP in patients infected by specific types of HPV. The quantitative analyses for pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated.

Results

Twenty‐six molecular epidemiological studies that recruited a total of 900 patients with SNIP were selected for the final meta‐analysis. The summary ORs indicated that HPV‐6 (OR: 2.02; 95% CI: 0.47–8.61; P = .343), HPV‐11 (OR: 0.86; 95% CI: 0.26–2.89; P = .806), and HPV‐6/11 (OR: 1.44; 95% CI: 0.59–3.53; P = .426) infections were not associated with the risk of malignant SNIP. However, the risk of malignant SNIP was increased in patients infected with HPV‐16 (OR: 8.51; 95% CI: 3.36–21.59; P < .001), HPV‐11/16 (OR: 7.95; 95% CI: 1.13–56.01; P = .038), HPV‐18 (OR: 23.26; 95% CI: 5.27–102.73; P < .001), and HPV‐16/18 (OR: 24.34; 95% CI: 5.74–103.18; P < .001).

Conclusions

This study found that patients infected with HPV types 16, 11/16, 18, and 16/18 were associated with an increased risk of malignant SNIP. However, patients infected with HPV types 6, 11, and 6/11 did not have a significant risk of malignant SNIP. Laryngoscope, 2020

View on the web

Effect of Sleep Surgery on C‐Reactive Protein Levels in Adults With Obstructive Sleep Apnea: A Meta‐Analysis

xloma.fota13 shared this article with you from Inoreader

Objectives/Hypothesis

To evaluate associations between sleep surgery and CRP (C‐reactive protein) levels in adults with obstructive sleep apnea (OSA).

Study Design

Meta‐analysis.

Methods

Two authors independently searched PubMed, Medline, EMBASE, and Cochrane review databases until July 2019. The keywords used were sleep apnea, OSA, sleep apnea syndromes, surgery, C‐reactive protein (CRP), and inflammatory markers. The effects of sleep surgery on CRP levels were examined using a random‐effects model.

Results

Nine studies with 277 patients were analyzed (mean age: 46.5 years; 92% men; mean sample size: 30.8 patients). The mean change in the apnea–hypopnea index (AHI) after surgery was significantly reduced by −21.1 (95% confidence interval [CI], −28.4 to −13.7) events/hr. Overall, sleep surgery resulted in a significant reduction of CRP levels in patients with OSA (standardized mean difference [SMD] = −0.39, 95% CI, −0.67 to −0.11). Patients with postoperative AHI reduction >20 events/hr achieved a greater reduction in CRP than those with AHI reduction <20 events/hr (SMD: −0.72 vs. −0.14, P for heterogeneity = .007). According to subgroup analysis, differences in the CRP levels after surgery were nonsignificant in the different countries (i.e., United States vs. other countries), CRP types (i.e., CRP vs. high‐sensitivity CRP), surgical procedures (i.e., pharyngeal surgery vs. other surgical procedures), and follow‐up period (i.e., <6 vs. >6  months).

Conclusions

Sleep surgery for OSA resulted in a significant reduction of CRP levels in adults. The beneficial effect of surgery on CRP levels is greater in patients with large improvement in OSA (i.e., AHI reduction >20 events/hr) after sleep surgery. Laryngoscope, 2020

View on the web

Botulinum Toxin Injection for Chronic Parotitis

xloma.fota13 shared this article with you from Inoreader

Objective

When conservative therapy fails for chronic parotitis, sialendoscopic treatment or surgical excision can be considered. However, these are financially costly or invasive. Thus, this study aimed to evaluate the clinical efficacy and safety of botulinum toxin (BTX) injection and to further analyze its effect on parotid gland function and volume using salivary single‐photon emission computed tomography (SPECT)‐computed tomography (CT).

Methods

This clinical trial has been registered in the Clinical Research Information Service, Republic of Korea. Fourteen patients with chronic parotitis received BTX injections into the parotid glands. Pain, postprandial pain, swelling, aesthetic discomfort, and salivary flow rate were assessed before injection, at 2 weeks, and at 1, 3, and 6 months. Salivary SPECT–CT was performed before injection and again 3 and 6 months after to assess the volume and uptake changes.

Results

All subjective symptoms decreased significantly until 1 month and then increased. However, at 6 months, all subjective symptoms were determined to be better than before injection. The unstimulated and stimulated salivary flow rate did not show a significant difference over time. No significant difference was noted in parotid gland volume or uptake on salivary SPECT–CT over time.

Conclusion

BTX injection can be an alternative treatment option for chronic parotitis.

Level of Evidence

4 Laryngoscope, 2020

View on the web

Colon Cancer Screening Should Start at Age 45

xloma.fota13 shared this article with you from Inoreader

Message:

By Dennis Thompson
HealthDay Reporter
WEDNESDAY, Oct. 28, 2020
Average folks should start being screened at age 45 to prevent colon cancer, five years earlier than is now recommended, the nation's top preventive medicine panel says.

The U.S. Preventive Services Task Force currently recommends that people aged 50 to 75 be regularly screened for colon cancer, one of a handful of cancers that can be prevented outright.

But new data suggests that screening earlier could save even more lives, said task force member Dr. Michael Barry, director of the Informed Medical Decisions Program in the Health Decision Sciences Center at Massachusetts General Hospital in Boston.

"We have epidemiologic data that the risks of colorectal cancer are increasing before age 50, particularly in that 45- to 49-year-old age group," Barry said.

Computer models suggest that about 25 colon cancer deaths are prevented for every 1,000 Americans between 50 and 75 who are screened, Barry said.

The earlier start is expected to prevent at least one more death per every 1,000 screened, Barry said.

Under the Affordable Care Act, health insurance companies are required to cover the full cost of any screening test recommended by the U.S. Preventive Services Task Force (USPSTF). The task force is an independent, volunteer panel of national experts in prevention and evidence-based medicine.

This guideline covers people at average risk for colon cancer, Barry said. People with factors that put them at higher risk -- for example, a strong family history of colon cancer -- might need to start screening even earlier, and should discuss it with their doctor.

Colon cancer almost always develops from precancerous polyps that form in the colon or rectum. These screening tests detect the presence of these polyps, so they can be removed before they turn into cancer.

Colonoscopy is the most widely known colon cancer screening method, but it's not the only one, Barry said.

"There are a whole group of tests that can reduce the risk that someone will die of colorectal cancer," he said.

For example, people can have their stool tested for the presence of tiny amounts of blood, which can indicate the presence of either cancer or polyps.

"That strategy, according to our modeling, can be about as good as colonoscopy in terms of reducing risk," Barry said.

The frequency a person undergoes screening would depend on the type of test they choose, Barry said. A person with a clean colonoscopy wouldn't have to be rechecked for a decade, while stool blood tests are often done every one to three years.

When the USPSTF last updated its colon cancer guidelines in 2016, it kept the age at which screening begins at 50 because they were concerned about the strength of the data supporting an earlier start, explained Robert Smith, senior vice president of cancer screening for the American Cancer Society.

The American Cancer Society updated its guidelines in 2018 to recommend that colon cancer screening start at age 45, he added.

"By 2018, we had new data that showed convincingly an increase in incidence in adults under the age of 50," Smith said. "The data has shown this is a birth cohort effect, and the incidence could be expected to continue to rise."

The USPSTF updates individual screening recommendations every four years, and "we anticipated when the task group updated their guidelines again in 2020 that they would likely reach the same decision," Smith said. "The good news now is that they've reached the same decision we reached two years ago."

"Now the public and physicians will have a set of recommendations that are for all practical purposes the same when it comes to when you should begin colorectal cancer screening," Smith said.

The bad news is that even now people who should get screened are not, Barry and Smith said.

"People do not routinely start screening at the age of 50," Smith said. "Usually the average age is in the mid-50s. That means during this period of rising incidence we are missing an opportunity to prevent colorectal cancer and detect it early."


SLIDESHOW
Colon Cancer: Symptoms, Signs, Screening, Stages
See Slideshow
"It looks like about 25% of eligible Americans have not been screened for colorectal cancer," Barry said. "That will lead to some colorectal cancer deaths that could be prevented, and that's always a tragedy."

The USPSTF is taking public comment on this draft recommendation from Oct. 27 through Nov. 23. The task force will consider all comments it receives, and release a final recommendation shortly afterward.

MedicalNews
Copyright © 2020 HealthDay. All rights reserved.

fb-mnet-default.jpg

Title: Colon Cancer Screening Should Start at Age 45
Category: Health News
Created: 10/27/2020 12:00:00 AM
Last Editorial Review: 10/28/2020 12:00:00 AM
View on the web

Botulinum Toxin Injection for Chronic Parotitis

xloma.fota13 shared this article with you from Inoreader

Objective

When conservative therapy fails for chronic parotitis, sialendoscopic treatment or surgical excision can be considered. However, these are financially costly or invasive. Thus, this study aimed to evaluate the clinical efficacy and safety of botulinum toxin (BTX) injection and to further analyze its effect on parotid gland function and volume using salivary single‐photon emission computed tomography (SPECT)‐computed tomography (CT).

Methods

This clinical trial has been registered in the Clinical Research Information Service, Republic of Korea. Fourteen patients with chronic parotitis received BTX injections into the parotid glands. Pain, postprandial pain, swelling, aesthetic discomfort, and salivary flow rate were assessed before injection, at 2 weeks, and at 1, 3, and 6 months. Salivary SPECT–CT was performed before injection and again 3 and 6 months after to assess the volume and uptake changes.

Results

All subjective symptoms decreased significantly until 1 month and then increased. However, at 6 months, all subjective symptoms were determined to be better than before injection. The unstimulated and stimulated salivary flow rate did not show a significant difference over time. No significant difference was noted in parotid gland volume or uptake on salivary SPECT–CT over time.

Conclusion

BTX injection can be an alternative treatment option for chronic parotitis.

Level of Evidence

4 Laryngoscope, 2020

View on the web

Phase I study of everolimus, letrozole, and trastuzumab in patients with hormone receptor-positive metastatic breast cancer or other solid tumors

xloma.fota13 shared this article with you from Inoreader

Purpose: Doublets of everolimus with letrozole or trastuzumab have demonstrated activity against HER2-positive breast cancer, suggesting that the triple combination can have synergistic anticancer activity. Experimental design: This first-in-human dose escalation study (NCT02152943) enrolled patients with hormone receptor-positive, HER2-positive (defined by amplification, overexpression or mutation)treatment-refractory advanced cancers to receive escalating doses (3+3 design) of daily oral letrozole (days 1-21), daily oral everolimus (days 1-21) and intravenous trastuzmab (day 1) every 21 days todetermine dose-limiting toxicities (DLTs) and maximum tolerated dose or recommended phase 2 dose (RP2D). Results: Total of 32 patients with hormone receptor-positive, HER2-positive (amplification, n=27; overexpression, n=1; mutation, n=4) advanced breast cancer (n=26) or other cancers (n=6) were enrolled. The most frequent grade greater than or equal to 3 adverse eve nts included hyperglycemia (n=4), anemia (n=3), thrombocytopenia (n= 2) and mucositis (n=2).DLTs included grade 3 mucositis and grade 4 neutropenia, and trastuzumab given as an 8-mg/kg loading dose on day 1 of cycle 1 followed by a 6-mg/kg maintenance dose on day 1 of subsequent cycles plus 10 mg everolimus daily and 2.5 mg letrozole daily every 21 days was declared as RP2D. Five breast cancer patients (4 with HER2amplification and 1 with HER2mutation) had partial responses. HER2amplification in circulating cell-free DNA at baseline was associated with shorter progression-free and overall survival durations (P<0.05). Conclusions: Everolimus, letrozole, and trastuzumab has a favorable safety profile and elicits encouraging signals of anticancer activity in patients with heavily pretreated hormone receptor-and HER2-positive advanced cancers.

View on the web

Nonalcoholic fatty liver disease: Reporting histologic findings in clinical practice

xloma.fota13 shared this article with you from Inoreader

Abstract

The role of liver biopsy in nonalcoholic steatohepatitis (NASH) has evolved along with the increased recognition of the significance of this disease, and the unmet medical need it presents. Drug development and clinical trials are rapidly growing, as are non‐invasive tests for markers of steatosis, inflammation, injury and fibrosis. Liver biopsy evaluation remains necessary for both drug development and clinical trials as the most specific means of diagnosis and patient identification for appropriate intervention.

This White Paper, sponsored by the American Association for the Study of Liver Disease NASH Task Force, is a focused review of liver biopsy evaluation in fatty liver disease in subjects with presumed NAFLD for practicing clinical hepatologists and pathologists. The goal is to provide succinct and specific means for reporting the histopathologic elements of NASH, distinguishing NASH from nonalcoholic fatty liver without steatohepatitis (NAFL), and from alcoholic steatohepatitis when possible. The discussion includes the special situations of NASH in advanced fibrosisor cirrhosis, and in the pediatric population. Finally, there is discussion of semiquantitative methods of evaluation of lesions of "disease activity" and fibrosis. Tables are present for scoring and a suggested model for final reporting. Figures presented highlight the histopathologic elements of NASH.

View on the web

Twin study of neonatal transient-evoked otoacoustic emissions

xloma.fota13 shared this article with you from Inoreader

Message:

Highlights

For the first time TEOAE heredity was studied in neonatal twins


A twin model suggests that TEOAE levels are largely inherited


Interaural and sex differences in TEOAEs are present from birth


The auditory function reflected in TEOAEs may be fully inherited


Abstract
Our knowledge of which physiological mechanisms shape transient evoked otoacoustic emissions (TEOAEs) is incomplete, although thousands of TEOAEs are recorded each day as part of universal newborn hearing-screening (UNHS). TEOAE heritability may explain some of the large TEOAE variability observed in neonates, and give insights into the TEOAE generators and modulators, and why TEOAEs are generally larger in females and right ears.

The aim was to estimate TEOAE heritability and describe ear and sex effects in a consecutive subset of all twins that passed UNHS at the same occasion at two hospitals during a six-year period (more than 30 000 neonates screened in total). TEOAEs were studied and TEOAE level correlations compared in twin sets of same-sex (SS, 302 individual twins, 151 twin pairs) and opposite-sex (OS, 152 individual twins, 76 twin pairs). A mathematical model was used to estimate and compare monozygotic (MZ) and dizygotic (DZ) intra-twin pair TEOAE level correlations, based on the data from the SS and OS twin sets.

For both SS and OS twin pairs TEOAE levels were significantly higher in right ears and females, compared to left ears and males, as previously demonstrated in young adult twins and large groups of neonates. Neonatal females in OS twin pairs did not demonstrate masculinized TEOAEs, as has been demonstrated for OAEs in young adult females in OS twin pairs. The within-twin pair TEOAE level correlations were higher for SS twin pairs than for OS twin pairs, whereas the within-pair correlation coefficients could not be distinguished from zero when twins were randomly paired. These results reflect heredity as a key factor in TEOAE level variability. Additionally, the estimated MZ within-twin pair TEOAE level correlations were higher than those for DZ twin pairs. The heritability estimates reached up to 100% TEOAE heritability, which is numerically larger than previous estimates of about 75% in young adult twins.

Key words
Twinheritabilityotoacoustic emissionear differencesex differenceneonatal
Abbreviations
DPOAEDistortion product otoacoustic emissionDZDizygoticMZMonozygoticOAEOtoacoustic emissionOHCouter hair cellOSOpposite-sexRMSRoot mean squareSDStandard deviationSFOAEStimulus-frequency otoacoustic emissionS/NSignal-to-noise ratioSOAESpontaneous otoacoustic emissionSSSame-sexTEOAETransient-evoked otoacoustic emissionUNHSUniversal newborn hearing-screeningWDRWeinberg's differential rule
1. Introduction
Otoacoustic emissions (OAEs) are sounds in the ear canal, caused by processes in the cochlea, directly associated with the hearing process (Kemp, 1978). OAEs comprise evoked and spontaneous OAEs (SOAE). The OAEs can be evoked by clicks or transients (transient-evoked OAE, TEOAE), one tone (stimulus-frequency OAE, SFOAE), or two nearby tones (the distortion product OAE, DPOAE). Furthermore, DPOAEs arise from nonlinear distortion, whereas low level TEOAEs, SFOAEs and SOAEs arise from linear reflection, that is, two different mechanisms in the organ of Corti (Shera and Guinan, 1999). Distortion emissions originate primarily from prestin-based mechanoelectrical transducer channels in outer hair cells (OHCs), and the distortion product is injected into the organ of Corti motion by OHC electromotility (Guinan, 2018; Ren and He, 2020; Shera and Guinan, 1999). Reflection emissions originate from the reflection of traveling-wave energy by minute irregularities along the cochlea, where the pea k of the travelling wave contributes most to the reflection (Guinan, 2018; Shera and Guinan, 1999). Even so, our understanding of the global system involved in shaping the OAEs is incomplete.

The OAE offers an objective, non-invasive measure of peripheral hearing sensitivity, where the TEOAE is widely used in clinical practice, partly due to its high intra-individual stability (Franklin, et al., 1992; Harris, et al., 1991; Johnsen and Elberling, 1982a; Marshall and Heller, 1996), and sensitivity in identifying minor cochlear hearing losses (e.g. Berninger, et al. (1995)). Thousands of TEOAEs are recorded each day worldwide as part of universal newborn hearing-screening (UNHS) programs.

While the intra-individual variability in TEOAE level in neonates is low, the neonatal inter-individual variability in TEOAE level is large (Berninger, 2007; Bray and Kemp, 1987). The causes for the large variability have only partly been explained. Sex differences (female > male) and ear asymmetries (right > left) exist in TEOAEs (Aidan, et al., 1997; Berninger, 2007; Kei, et al., 1997; Thornton, et al., 2003), although these effects may not be observed in samples smaller than approximately 500 neonates (Cassidy and Ditty, 2001; Johnsen, et al., 1988; Khalfa, et al., 1997).

The sex differences in OAEs have been attributed to the sex differences in exposure to androgens during prenatal development (McFadden, 1993b; McFadden, 2009). Support for this theory can be found in studies of hyenas (McFadden, 2008; McFadden, et al., 2006b), sheep (McFadden, et al., 2009), and rhesus monkeys (McFadden, et al., 2006a), where prenatal hormone levels were manipulated. An explanation suggested for the ear difference in OAEs is that efferent inhibition may be relatively less in right ears (McFadden, 1993b), as the major efferent neural pathway synapse on OHCs. That the olivocochlear systems are involved in modulating the OAEs has been demonstrated (Guinan, 2006; Guinan, 2018; Veuillet, et al., 1991). However, it is yet unknown where in the auditory system ear and sex differences stem from, although the main effects presumably do not arise in the outer and middle ear. McFadden (1993b) argued that ear and sex differences have been demonstrated at the level of the cochlea, but not in the outer and middle ear, also supported by results of no ear and sex effect in electrically constant TEOAE peak stimulus levels in the outer ear canal (Berninger, 2007), in a study where ear and sex effects were demonstrated for TEOAEs.

The effect of inheritance of TEOAE levels could contribute to our understanding of which structures are involved in shaping TEOAEs and the large inter-individual TEOAE variability. One previous study exists of TEOAE heredity, where it was estimated that about 75% of the variation in TEOAE level could be attributed to heredity, by comparing monozygotic (MZ) and dizygotic (DZ) young adult twins (McFadden, et al., 1996). The percentage not explained by heredity was suggested to be caused by differential exposure to sex hormones prenatally (McFadden, et al., 1996). In support of a prenatal masculinization effect a twin testosterone transfer effect was indicated for SOAEs (McFadden, 1993a; McFadden and Loehlin, 1995), and a similar effect was observed for TEOAEs (McFadden, et al., 1996), although not statistically significant. Young adult female twins in opposite-sex (OS) twin pairs had half as many SOAEs, compared to female twins in same-sex (SS) twin pairs, corresponding to the prevalen ce in young adult males (McFadden, 1993a; McFadden and Loehlin, 1995). An influence of sex hormones on the cochlear amplifier in humans is not unlikely, as drugs like quinine and salicylate influence the presence and magnitude of OAEs (Alvan, et al., 2017; Johnsen and Elberling, 1982b), including TEOAEs (Berninger, et al., 1995; Karlsson, et al., 1991), and presumably block the cochlear amplifier and the motor protein prestin (Alvan, et al., 2017). However, more research is needed in order to link the effect of androgens on the cochlear amplifier to a prenatal period in humans. The twin testosterone transfer effect was observed in twins that were students at the university, and different hormones can influence TEOAEs in young adults (see e.g. Burke, et al. (2020)). Additionally, a postnatal surge, sometimes referred to as "mini puberty" (Hines, et al., 2015), may also have effects on TEOAE levels. During this period (peak 1-2 months after birth) urine testosterone levels are not ably higher in males than in females, while no testosterone sex difference exists in newborns (7 days of age) and in 6-month-old infants (Hines, et al., 2015; Lamminmäki, et al., 2012).

We are not aware of any studies of TEOAE inheritance in neonatal twins, where the environmental factors between two twins in a pair is minimal. In previous studies of OAE inheritance in young adult twins, differences in environmental exposure needed to be considered (McFadden and Loehlin, 1995; McFadden, et al., 1996), and heritability models taking environment into account have limitations. More research in large groups of neonates is needed, due to the incomplete understanding of the large inter-subject variability in TEOAE strength, as well as ear and sex differences. Specifically, the study of heredity in neonates, in combination with advancements in cochlear mechanics, can improve our understanding of OAE generation and modulation.

The aim was to study ear and sex differences in a consecutive subset of all twins that passed the UNHS program in both ears at the same test occasion at two hospitals during a six-year period (more than 30 000 newborns screened). The purpose was to estimate TEOAE heredity, and to study ear and sex differences in TEOAE level in twins and non-twins. We demonstrate the presence of ear and sex differences in TEOAE from the neonatal period, and that the individual differences in neonatal TEOAEs may be fully inherited. How these results may influence the current understanding of the auditory system involved in shaping OAEs is discussed.

2. Material and Methods
2.1. Study design
TEOAEs were recorded from more than 30 000 neonates (98% of all newborns) at Karolinska University Hospital, Huddinge and Södertälje Hospital, Södertälje, Sweden, from November 1998 until the end of 2004 (Berninger, 2007; Berninger and Westling, 2011). The bedside UNHS was based on multiple TEOAE recordings to enhance the specificity of the screening program (Berninger, 2014). Off-line analysis was performed on the entire TEOAE levels of all twin sets that passed UNHS in all four ears at the same test occasion, and had valid data on ear tested, sex, name, and test date.

To estimate heredity, intra-twin pair TEOAE level relationships were studied. Specifically, the TEOAE intra-twin pair relationships were compared for SS pairs and OS pairs, and estimates of MZ and DZ relationships were compared. TEOAE relationships also were studied for twins that were randomly paired, as a non-heredity and sex-effect control. Finally, broad heritability was estimated with the Falconer's formula based on the difference between the estimated MZ and DZ correlations (Falconer and Mackay, 1996). All non-twins that passed the UNHS in both ears, with valid data on ear tested, sex, name, and date of test, were used as a non-twin control group (n = 21 199 individuals).

This study was approved by the Swedish Ethical Review Authority (no: 2019–03826).

2.2. Transient-evoked otoacoustic emissions (TEOAEs)
TEOAEs were recorded in the non-linear quickscreen default mode with ILO 288 (Otodynamics Ltd., program version 5.6 y, time window: 3.0-12.8 ms, click rate 78 Hz, for details on the non-linear stimulus paradigm see Kemp, et al. (1990)). All the TEOAE stimulus and response levels were recorded at the probe-tip in the outer ear canal using an electrically constant stimulus, corresponding to a typical stimulus level of 81.8 dB SPL peak (Berninger, 2007).

TEOAE pass criteria included: ≥ 70% whole wave reproducibility, signal-to-noise ratio (S/N) ≥ 3 dB in at least three of the upper four wide-frequency bands provided by the ILO instrument (center frequencies from about 1500 to 4000 Hz, ± 400 Hz bandwidth), and ≥ 50 sweeps.

The entire TEOAE level was calculated as the root mean square (RMS) value of the time signal. A sweep consisted of two sets of non-linear responses in response to four 80-μs click stimulations. Whole-wave reproducibility was calculated as the correlation coefficient of interleaved non-linear responses.

TEOAEs from all except three twin pairs (i.e., 99%) were obtained at the first TEOAE test occasion, which was typically performed at postnatal day 3 (Berninger and Westling, 2011), during a period when it was typical for the mothers to stay at least three days at the hospital after delivery. The left ear was tested first in 58% of all neonates that were screened during the six-year period (Berninger and Westling, 2011). The twins were included in a consecutive manner, which should be important, as TEOAEs have been demonstrated to be larger in the ear tested first (Thornton, et al., 2003).

2.3. Data files and data analysis
A Matlab program was developed for retrieving data from the TEOAE files generated by ILO 288 (file extension: dta) and cross-checking pass criteria. It was also used for computing response levels in non-overlapping half-octave frequency bands, geometrically centred at 707, 1000, 1414, 2000, 2828 and 4000 Hz, where the four upper frequency bands were used for the S/N pass criteria.

2.4. Subjects
In total, 642 individual twins (321 pairs) were identified in a clinical database of all newborns that had passed UNHS at Karolinska University Hospital, Huddinge and Södertälje Hospital, Södertälje, Sweden, during the six-year period. Out of the identified twins, the custom-made Matlab program identified 454 individual twins (227 pairs) that passed with the strict inclusion criteria, i.e., passed bedside UNHS in all four ears at the same test, and had datafiles that included information on ear tested, sex, name and test date.

In all, 227 twin pairs were included, 151 SS twin pairs (66.5%) and 76 OS twin pairs (33.5%). Of all the 454 individual twins, 51.1% were male. Of the SS twin pairs, 78 twin pairs were male (51.6%). The first-born and second-born twins are henceforth denoted twin 1 and twin 2.

The non-twin comparison group consisted of 21 199 neonates (50.3% males), derived from the material of all >30 000 screened neonates (Berninger (2007)). The inclusion criteria comprised data on ear tested, sex, name and test date, whereafter the twins in the material were excluded based on the file name. The sex inclusion criterion was somewhat different for the twin group and the control group, due to the sample size. The sex information was manually typed into the ILO-program, and for the large non-twin group inclusion was strictly based on checking that of all but the last typed letter corresponded to the Swedish word for boy or girl. In the smaller twin group, the sex of the individual twins (n = 454) could be verified manually, so that the twins with, e.g., a "p" for "pojke" (i.e., Swedish word for boy) or the Swedish word for "twin" instead of the sex information could be included in the analysis. Typing errors of information on sex were a main reason for exclus ion of TEOAEs in the non-twin group. Other main reasons for exclusion were less than 70% whole-wave reproducibility and less than 3 dB S/N in three of the four upper half-octave frequency bands (i.e., 1414, 2000, 2828 and 4000 Hz), especially in the twin group, as all four ears were excluded if one ear did not fully correspond to the inclusion criteria.

2.5. Estimated monozygotic (MZ) and dizygotic (DZ) intra-twin pair correlations
A mathematical model was used to estimate and compare MZ and DZ intra-twin pair TEOAE level correlations, based on the data from the SS and OS twin pairs. The model uses Weinberg's differential rule (WDR) (Fellman and Eriksson, 2006), which was created for estimation of MZ and DZ twinning rates in twin studies. WDR has been demonstrated to be robust despite its simplicity and gives reliable results when official birth registers are analysed (Fellman and Eriksson, 2006). WDR is implicitly based on the assumptions that the probability of a male equals the probability of a female in the entire twin material (our material 51.1% males), and that there is independence between the sexes in DZ twin pairs, which is true for all OS twin pairs (same number of males and females) (Fellman and Eriksson, 2006). For DZ twin pairs of SS we based our assumption on independence between the sexes on the SS twin pairs (our twin material: 51.6 % male SS twin pairs, close to 50%).

The OS twin pairs are DZ. Hence, the TEOAE variance and correlation coefficient for the DZ twins was calculated from the OS twin pairs.

According to WDR the rate of DZ twinning is twice the rate of twin maternities in which the twins are of OS (NDZ = 2NOS, our set: nDZ estimated = 2nOS = 152). The MZ twinning rate is the difference between the rates of SS and OS twin pairs (NMZ = NSS – NOS, our set: nMZ estimated = nSS - nOS = 151-76 = 75). Accordingly, the number of DZ pairs in the SS set should then approximately be equal to the estimated number of MZ twin pairs (our set: nDZ estimated in SS group = nSS – nMZ estimated = 151-75 = 76 ≈ nMZ estimated). Additionally, based on the 50% probability of a MZ twin set (75/151 = 0.50), and 50% probability of a DZ twin set (76/151 = 0.50), for a random twin pair in the SS twin sets, MZ twin's TEOAE variance was estimated as:
(1)
where P is probability, and σSS are estimated from the TEOAE levels of the SS set, and σDZ from the TEOAE levels of the OS set.

Then, the correlation coefficient (ρss) was calculated according to:
(2)
where P is probability, and X is for twin 1 and Y for twin 2 in each twin set. It is assumed that the standard deviation is equal for twin 1 (x) and twin 2 (y) in the population. Thus:
(3)

From equations 1 and 3 the correlation coefficient for the MZ twins is estimated as:
(4)
where the variance for the SS twins was estimated as 0.5(variance for twin 1 + variance for twin 2) from the SS set, and the variance for the DZ twins was estimated as 0.5(variance for twin 1 + variance for twin 2) from the OS set.

2.6. Estimate of heritability
Falconer's formula was used to calculate broad heritability (Falconer and Mackay, 1996):
(5)
where the MZ and DZ intra-twin pair correlation coefficients (rMZ and rDZ) are estimated from equations 1-4. Falconer's formula is twice the difference in correlation between MZ and DZ twins, which originate from MZ twins sharing all the same genes, whereas DZ twins normally share half their genes, so MZ twins are on average twice as genetically similar as DZ twins (Falconer and Mackay, 1996).

2.7. Twins in randomized pairs
The TEOAE levels for randomly paired twins were compared, as a control that differences in correlations were driven by heredity (between OS and SS pairs, and estimated DZ and MZ correlations). In the dataset of the 227 twin pairs, the TEOAE level for twin 1 in twin pair n was compared to that of twin 2 in twin pair n+1 (twin 2 from pair 1 was paired with twin 1 in the final pair).

Additionally, correlations were studied for twins that were randomly paired under condition that their randomly assigned co-twin was of the same sex as their biological co-twin (i.e., randomization with stratification for sex). The analysis was performed to control if sex may be a confounding variable when comparing the SS and OS twin groups. Accordingly, the dataset was divided into four subsets: 1) male SS-pairs, 2) female SS-pairs, 3) OS pairs with a male twin 1, and 4) OS pairs with a female twin 1. Within each subset twin 1 from pair n was paired with twin 2 from pair n+1 (twin 2 from twin pair 1 was paired with twin 1 in the final pair in each subset). Thereafter subsets 1 and 2 were combined into a SS group and subsets 3 and 4 were combined into an OS group for analysis.

2.8. Statistical analysis
All the statistical analyses were performed with Statistica version 13 (Statsoft Inc., USA).

One-way ANOVA was used when comparing the TEOAE level for the SS, OS and non-twin group. Factorial ANOVA was used for ear and sex effects on TEOAE level. Moreover, paired and unpaired t-tests were used for the study of sex and ear differences on data that reflected a Gaussian distribution. The average TEOAE level for each twin pair and ear was used, to avoid violation of independent observations (same method as used by McFadden and Loehlin (1995)).

Non-parametric tests were used for data that reflected a non-Gaussian distribution, e.g., the stimulus levels. Wilcoxon's matched-pairs test was used for dependent samples, and the Mann-Whitney U test for independent samples. Moreover, for data that reflected non-Gaussian distributions, medians are presented instead of means, and in Table 1 medians and means.

Table 1. TEOAE levels classified by group, ear, and sex (mean, standard deviation (SD), median, quartiles, and non-outlier range (coefficient 1.5), and n denotes number of neonates).

Mean SD Median 25-75% Min-Max n
All twins Ear-average All 19.6 5.1 19.6 16.1-23.1 7.1-32.4 454
♀ 20.4 4.6 20.0 17.2-23.7 8.5-32.4 222
♂ 18.9 5.6 18.7 14.4-22.7 7.1-32.1 232
Left All 18.9 5.6 18.8 14.9-22.5 5.8-32.4 454
♀ 19.7 5.0 19.6 16.5-22.9 6.6-32.4 222
♂ 18.0 6.0 17.6 12.8-22.3 5.8-32.1 232
Right All 20.4 5.6 20.8 16.5-24.3 6.2-34.5 454
♀ 21.1 5.0 21.2 17.4-24.6 8.0-32.4 222
♂ 19.7 6.0 20.2 15.3-24.2 6.2-34.5 232
SS Ear-average All 19.6 5.2 19.5 15.9-23.3 8.5-32.4 302
♀ 20.2 4.6 19.8 17.2-23.8 8.5-32.4 146
♂ 19.0 5.6 18.6 14.3-22.7 8.6-32.1 156
Left All 18.8 5.7 18.8 14.6-22.5 5.8-32.4 302
♀ 19.5 5.1 19.4 15.8-22.5 6.6-32.4 146
♂ 18.1 6.1 17.5 12.6-22.6 5.8-32.1 156
Right All 20.4 5.5 20.9 16.5-24.1 6.5-34.5 302
♀ 20.9 5.2 21.3 17.3-24.5 8.0-32.4 146
♂ 19.9 5.8 20.1 15.7-23.8 6.5-34.5 156
OS Ear-average All 19.7 5.1 19.7 16.5-22.9 7.1-31.2 152
♀ 20.7 5.6 20.5 17.7-23.5 10.8-31.2 76
♂ 18.6 4.4 19.3 14.6-22.0 7.1-29.3 76
Left All 19.1 5.3 18.8 15.4-22.3 7.1-32.1 152
♀ 20.2 5.6 19.6 17.1-23.4 11.1-32.1 76
♂ 17.9 4.7 17.7 14.2-21.9 7.1-31.7 76
Right All 20.3 5.6 20.7 16.4-24.7 6.2-31.4 152
♀ 21.3 6.3 21.0 17.7-24.6 10.5-31.4 76
♂ 19.3 4.7 20.2 14.7-24.8 6.2-29.7 76
Non-twin Ear-average All 18.9 4.9 19.0 15.5-22.3 5.4-32.3 21 199
♀ 19.5 4.9 19.6 16.2-22.9 6.1-33.0 10 526
♂ 18.3 4.8 18.4 15.0-21.6 5.4-31.5 10 673
Left All 18.4 5.3 18.4 14.7-22.1 3.7-33.1 21 199
♀ 19.0 5.3 19.1 15.3-22.8 4.3-33.7 10 526
♂ 17.7 5.2 17.8 14.1-21.3 3.4-32.0 10 673
Right All 19.5 5.3 19.5 15.8-23.1 4.9-34.0 21 199
♀ 20.0 5.3 20.1 16.4-23.7 5.4-34.6 10 526
♂ 18.9 5.2 19.0 15.3-22.5 4.6-33.3 10 673
SS = Same-sex twin group. OS = Opposite-sex twin group.


Pearson's correlation coefficient was calculated for co-twins' TEOAEs to quantify twin resemblance in TEOAE. The differences between correlation coefficients were computed using Fisher's r-to-z transform with an unpaired two-tailed t-test.

Additional effect sizes were calculated for the main TEOAE sex and ear differences, as well as the difference between estimated MZ and DZ intra-twin pair correlations. The effect sizes for sex and ear effects were calculated as the difference between two means divided by the square root of the weighted mean of the two variances (Cohen's d), where effect sizes of 0.2, 0.5, and 0.8 are viewed as small, medium and large, respectively (Cohen, 1992). The effect size for the difference between correlations was calculated as the difference between two z-values after using the Fisher's r-to-z transform, where effect sizes of 0.1, 0.3, and 0.5 are viewed as small, medium and large, respectively (Cohen, 1992).

Bootstrapping was used to estimate the variance of the heritability estimates. The input data were the ear-average TEOAEs of the included 227 twin pairs, divided into the SS and OS groups, as in the main heritability analysis. Then, the number of twin pairs found before we applied our strict inclusion criteria, i.e., 321 pairs, was used as the number of slots available for resampling from the 227 twin pairs' data. The resampling was performed 10 000 times with different random samples of twin pair data. Variances and correlations were calculated for each resample (Eq. 1-4), and the 10 000 calculations of H2 were used to estimate the broad heritability variance.

3. RESULTS
3.1. TEOAE stimulus and response parameters
No lateral asymmetries or sex differences existed in stimulus levels. The median interaural peak stimulus level difference was 0.0 dB (n = 454) and 0.0 dB (n = 21 199) for the individual twins and non-twins, respectively. The sex difference (females-males) in median peak stimulus level was 0.0 dB (n = 454) for the individual twins, and -0.1 dB (n = 21 199) for the non-twins. Moreover, twins and non-twins displayed no significant difference in stimulus levels (U = 2234506, p = .06), with a median of 81.9 dB SPL peak for the twins (n = 227; twin pair average), and 81.5 dB SPL peak for the non-twins (n = 21 199). No significant correlation was found for TEOAE level as a function of (the electrically constant) stimulus level in twins (r = -.02, p = .71, n = 454 individual twins), indicating that the variability in TEOAE levels could not be explained by the small variability in stimulus levels (3.1 dB inter-quartile range).

The median TEOAE levels were 19.4 dB SPL (n = 227; twin pair average) for the twins and 19.0 dB SPL (n = 21 199) for the non-twins, with a significant difference between the groups (U = 2221052, p = .045) (see Table 1 for TEOAE level by group, ear and sex). A median of 56 sweeps were used (n = 908 twin ears), and the median S/N was 9.6 dB (non-outlier range: 4.5-25.2 dB, n = 908 twin ears). Moreover, the whole-wave reproducibility was high (median = 94%, n = 908 twin ears).

3.2. TEOAE ear and sex differences
The twins showed statistically significant effects of ear (F(1, 904) = 17.1, p < .0001) and sex (F(1, 904) = 17.5, p < .0001) on TEOAE level, with larger TEOAEs in right ears and females (Table 1).

The mean TEOAE levels were 1.5 dB larger in right compared to left ears in twins (p < .0001, n = 227; twin pair average), as compared to the average 1.1 dB difference for non-twins (p < .0001, n = 21 199). Likewise, significant TEOAE interaural differences (right > left) existed in the subgroups of OS twin pairs (p = .0005, n = 76; twin pair average) and SS-twin pairs (p < .0001, n = 151; twin pair average). For further ear comparisons see Table 1, although the TEOAE levels are shown for each neonate, not the twin pair averages used for statistical purposes. No significant difference was found when comparing the interaural difference between twins in SS pairs (mean = 1.6 dB), twins in OS pairs (mean = 1.2 dB), and non-twins (mean = 1.1 dB) (F(2, 21 650) = 2.8, p = .055) (Table 1).

Female twins (n = 222 individuals) showed 1.5 dB larger mean TEOAE levels (ear-average) compared to male twins (n = 232 individuals) (p = .001), similar to the 1.2 dB sex difference for non-twins (p < .0001, n = 21 199) (Table 1). In the subgroups, females in OS twin pairs showed 2.1 dB larger mean TEOAE levels (ear-average) compared to their male co-twins (p = .002, n = 76 twin pairs, 152 individual twins) (Table 1). Moreover, female SS-pairs (n = 73 pairs, 146 individual twins) showed 2.1 dB larger median TEOAE levels (ear-average) compared to male SS-pairs (n = 78 pairs, 156 individual twins) (U = 2273, p = 0.03).

Cohen's d for the effect of sex on ear average TEOAE was 0.29, 0.23, 0.42, and 0.25 for the groups of all twins, SS, OS, and non-twins, respectively (see Table 1). Corresponding figure for the effect of ear was 0.27, 0.29, 0.22, and 0.21, respectively.

To examine whether a twin testosterone effect existed, the subgroups were analyzed for males and females separately. A significant difference between the OS, SS and the non-twin group was found for females in mean TEOAE level (F(2, 10745) = 3.6, p = .03), but not for males (F(2, 10902) = 1.5, p = .22) (Figure 1). However, no significant ear-average TEOAE level difference was found between female twins in OS-pairs (20.7 dB SPL, n = 76 females), and female twins in SS-pairs (20.2 dB SPL, n = 146 females) (p = 0.41), indicating no twin testosterone effect. The statistically significant difference for females revealed by ANOVA was most likely driven by female twins in OS-pairs (n = 76 females) having 0.9 dB larger median TEOAEs than female non-twins (n = 10526 females) (U = 346743, p = .045) (Figure 1).

Figure 1
Download : Download high-res image (164KB)Download : Download full-size image
Figure 1. Mean TEOAE levels (ear-average) categorized by sex for twins in opposite-sex (OS) twin pairs (n = 76 males, n = 76 females), twins in same-sex (SS) twin pairs (n = 156 males, n = 146 females), and for non-twins (n = 10673 males, n = 10526 females). Vertical bars denote 95% confidence intervals, *p < 0.05, **p < 0.01, ***p < 0.0001. The sex difference within OS-pairs was calculated with a paired t-test, the OS-twin vs. non-twin comparison with a Mann-Whitney U test, and the sex difference in non-twins with an unpaired t-test.

Post-hoc analysis of ear and sex effects in a random sample of non-twin neonates with the same sample size as the twins (n = 454 individuals, 2.1% of the non-twin group) also was performed due to the statistically significant effects in the twin group. A significant effect of ear (p < .0001, n = 454 non-twins) and sex (p = 0.02, n = 454 non-twins) existed in TEOAE levels (50% males in the random sample chosen by Statistica).

3.3. TEOAE correlations for opposite-sex (OS) and same-sex (SS) twin pairs
The twins in SS pairs showed a significantly larger mean TEOAE intra-twin pair correlation coefficient of r = .52 (n = 151 pairs, 302 individual twins), compared to the r = .27 (n = 76 pairs, 152 individual twins) for the twins in OS pairs (p = 0.04) (Table 2). Depending on ear under comparison, the TEOAE correlation coefficients varied between r = .37-.48 (n = 151 pairs, 302 individual twins) in SS-pairs, and r = .19-.30 (n = 76 pairs, 152 individual twins) in OS-pairs (Table 2), thereby reflecting the genetic contribution to the TEOAE levels, as ≈ 50% of SS twin pairs are MZ. The intra-twin pair TEOAE correlation between the left ear for twin 1 and the right ear for twin 2 (L1-R2) and the right ear for twin 1 and the left ear for twin 2 (R1-L2) were combined into a single opposite-ear comparison in Table 2 (L1-R2 + R1-L2). The opposite-ear comparison demonstrated larger intra-twin pair resemblance for SS twins as compared to OS twins (Table 2). It should be noted that both ears of both twins (dependent observations) were included in the L1-R2 + R1-L2 comparison, and if studied separately the L1-R2 condition also showed a statistical difference between the groups (SS twins: r = .43, n = 151 pairs, 302 individual twins vs OS twins: p = .16, n = 76 pairs, 152 individual twins; difference p = .04), although the R1-L2 condition did not reach statistical significance (SS twins: r = .47, n = 151 pairs, 302 individual twins vs OS twins: p = .24, n = 76 pairs, 152 individual twins; difference p = .06).

Table 2. TEOAE intra-twin pair correlations for twin pairs of same-sex (SS, n = 151 pairs, 302 individual twins) and opposite-sex (OS, n = 76 pairs, 152 individual twins). The analysis included both left and right ears for the L1-R2 + R1-L2 comparison (SS, n = 302 ears; OS, n = 152 ears). The differences between correlation coefficients were calculated using Fisher's r-to-z transform with a two-tailed t-test.

Twin 1 vs. Twin 2 SS OS rSS vs. rOS
r p r p p
L-L .48 < .0001 .30 .01 .14
R-R .37 < .0001 .24 .03 .32
L1-R2 + R1-L2 .42 < .0001 .19 .02 .01
Ear-average TEOAE-Ear-average TEOAE .52 < .0001 .27 .02 .04
L=left ear, R=right ear. Twin 1 was born before Twin 2.


3.4. TEOAE correlations for randomly paired twins
The correlation coefficients of TEOAE levels between ears in randomized pairs were close to zero; r = .03 for left ears (L-L), r = .00 for right ears (R-R), and r = .06 (L1-R2) and r = -.04 (R1-L2) for opposite ears (n = 227 non-biological pairs, 454 individual twins) (L1-R2 + R1-L2: r = -.01, n = 454 individual twins, n = 908 individual ears).

3.5. Within-subject TEOAE correlations
The interaural TEOAE correlation coefficients were similar for twins (r = .71, p < .0001, n = 454 individual twins), non-twins (r = .70, p < .0001, n = 21 199), twins in SS-pairs (r = .69, p < .0001, n = 302 individual twins), and twins in OS-pairs (r = .77, p < .0001, n = 152 individual twins) (Table 4).

3.6. Estimated TEOAE heritability
The estimated intra-twin pair MZ correlation based on ear-average TEOAE level (r = .77, p < .0001, n = 75 pairs, 150 twins) was significantly higher (p < .0001) than the estimated intra-twin pair correlation for DZ twins (r = .27, p = .03, n = 76 pairs, 152 twins) (Table 5). Depending on ear under comparison, the estimated intra-twin pair DZ correlations varied between r = .19-.30 (n = 76 pairs, 152 twins), compared to r = .51-.68 for the estimated MZ correlations (n = 75 pairs, 150 twins) (Table 5). Separate opposite-ear comparisons also demonstrated statistical significance between estimated MZ and DZ correlations, i.e., the L1-R2 comparison (estimated MZ: r = .67, n = 75 pairs, 150 twins vs DZ: r = .16, n = 76 pairs, 152 twins; difference p = .0001) and R1-L2 comparison (estimated MZ: r = .68, n = 75 pairs, 150 twins vs DZ: r = .24, n = 76 pairs, 152 twins; difference p = .0006).

The effect size for the difference between the estimated intra-twin pair MZ and DZ correlations was 0.74, indicating a large effect (0.5 = large).

The estimated broad heritability based on the average TEOAE level of each twin was H2 = 1.0, i.e. 100% heredity. When comparing specific ears, the estimated heredity of TEOAE level was at maximum for opposite ears, H2 = .95, while the corresponding heritability estimate for same ears was H2 = .59 (Table 5).

The bootstrapping analysis based on ear-average TEOAEs demonstrated a median of approximately 1 and variance of 0.33 for H2. Of all H2 estimates, 75% exceeded 0.69.

When the randomized pairs for the SS and OS groups were compared (randomization with stratification for sex within each subgroup), the correlation coefficients of TEOAE levels were likewise close to zero, with no significant difference between the groups (Table 3). The results support the interpretation that the comparatively high intra-twin pair correlation coefficients for the SS group in Table 2 were due to heredity, not because the twins were of SS (as compared to OS). This was also true for separate L1-R2 comparisons (SS: r = .06, n = 151 pairs, 302 individual twins vs OS: r = -.08; n = 76 pairs, 152 individual twins; difference: p = .33) and R1-L2 comparisons (SS: r = .08, n = 151 pairs, 302 individual twins vs OS: r = .05, n = 76 pairs, 152 individual twins; difference: p = .83).

Table 3. TEOAE intra-pair correlations for randomly paired twins shown for the twin groups of same-sex (SS, n = 151 non-biological pairs) and opposite-sex (OS, n = 76 non-biological pairs), where the randomly paired co-twin is of the same sex as their biological co-twin. The analysis included both left and right ears for the L1-R2 + R1-L2 comparison (SS, n = 302 ears; OS, n = 152 ears). The differences between correlation coefficients were calculated using Fisher's r-to-z transform with a two-tailed t-test.

Twin 1 vs. Twin 2 SS OS rSS vs. rOS
r p r p p
L-L .09 .27 -.01 .94 .48
R-R .07 .40 .05 .66 .89
L1-R2 + R1-L2 .05 .42 -.03 .74 .44
Ear-average TEOAE- Ear-average TEOAE .09 .28 .01 .97 .58
L=left ear, R=right ear. Twin 1 was born before Twin 2.


4. Discussion
The individual differences in neonatal TEOAE levels are largely inherited. Our estimates of heredity based on the TEOAEs of neonatal twins that passed UNHS suggest that typical TEOAE levels may be up to 100% inherited, in comparison to the previous heredity estimate in young adults of about 75% (McFadden, et al., 1996).

The heritability results are consistent, i.e., estimated MZ within-pair TEOAE level relationships were significantly closer than estimated TEOAE level relationships for DZ pairs (Table 5), twins in SS-pairs showed significantly closer within-pair TEOAE level resemblance than twins in OS-pairs (Table 2), and randomly paired twins showed no TEOAE level resemblance (Table 3). Further supporting a large influence of heredity, the estimated MZ within-pair TEOAE level relationships (Table 5), were as close as the within-subject TEOAE level relationships for twins and non-twins (Table 4). The heredity results show that neonatal TEOAEs are nonrandom events. Moreover, that TEOAE levels are very similar within-twin pairs is noteworthy in light of the generally large neonatal variability (Berninger, 2007; Bray and Kemp, 1987) and high stability of the TEOAE (Franklin, et al., 1992; Harris, et al., 1991; Johnsen and Elberling, 1982a; Marshall and Heller, 1996). Moreover, the estimated 100% hered ity based on the ear-average TEOAE indicates that both the generation and the congenital modulation, e.g. reflected in ear and sex differences in TEOAE level, may be fully inherited.

Table 4. TEOAE interaural correlation coefficients for all twins, twins in same-sex (SS) pairs, twins in opposite-sex (OS) pairs, and non-twins.

Group n r p
All twins 454 .71 < .0001
SS 302 .69 < .0001
OS 152 .77 < .0001
Non-twins 21 199 .70 < .0001
In support of a large influence of heredity on TEOAE, genetically coded structures involved in cochlear mechanical motions have been demonstrated to affect TEOAEs, e.g., based on knockout mice studies. TEOAEs and SFOAEs were enhanced, and SOAEs were more prevalent in mutant mice without Hensen's stripe and the protein CEACAM16. Hensen's stripe is a prominent feature in the basal two-thirds of the tectorial membrane situated above the stereocilia of the hair cells (Cheatham, et al., 2014). Besides cochlear structures, it is also well-known that the medial olivocochlear (MOC) efferents can modulate TEOAE levels (e.g., Guinan, 2018, for a recent review), as MOC neurons have myelinated axons that synapse on OHCs. However, it is not yet established whether the efferents are responsible for ear (and/or sex) differences in TEOAE level, as suggested by McFadden (1993b).

Our results reveal statistically significant effects of ear (right 1.5 dB > left) and sex (female 1.5 dB > male) on twin TEOAE levels, that previously have been demonstrated in adult twins (McFadden, et al., 1996), and large groups of neonates (Aidan, et al., 1997; Berninger, 2007; Kei, et al., 1997; Thornton, et al., 2003). The TEOAEs were recorded in 99% of the twins at the first TEOAE occasion (typically on postnatal day 3), before the second surge of testosterone (Hines, et al., 2015), confirming that sex and ear differences in TEOAE are present from birth. The differences may not be of clinical importance due to the relatively large variability and 1.5 dB in size (Table 1), which is also reflected in the small effect sizes indicated by Cohen's d for the twins and non-twins (0.21-0.42). However, the effects are noteworthy in the understanding of the TEAOE, e.g., indicating a prenatal origin of these differences. Effects of both ear and sex on TEOAEs have typically not been observ ed in samples of less than 500 neonates before, from no effect in 100 neonates (Johnsen, et al., 1988), to only an effect of sex in 350 neonates (Cassidy and Ditty, 2001), to only an effect of ear in 483 preterm neonates (Khalfa, et al., 1997). One explanation for our observation of statistically significant effects in the twin material could be strict inclusion criteria and good recording conditions with a median TEOAE S/N of 9.6 dB, and that we included all subjects that were born during a six-year period that corresponded to the inclusion criteria. Significant effects of ear and sex in a random sample from the non-twin group, as large as the twin group (n = 454 individuals, 2.1% of the non-twin group; post hoc analysis), support this hypothesis.

Our results on TEOAEs did not support the twin testosterone transfer hypothesis (McFadden, 1993a; McFadden and Loehlin, 1995; McFadden, et al., 1996), as the females in OS twin pairs showed no indication of masculinization of their auditory systems a few days after birth (Figure 1), in a larger subject group than that of young adult twins studied by McFadden, et al. (1996). The idea of twin testosterone transfer originates from research on male and female traits in litter-bearing rodents (Clemens, et al., 1978; Kinsley, et al., 1986; vom Saal, 1989; vom Saal and Bronson, 1980). For example, female rats positioned between two males during gestation have demonstrated atypically frequent mounting behavior, that was significantly reduced in response to prenatal anti-androgen treatment (Clemens, et al., 1978). Thus, it is not unreasonable to expect that females in OS-pairs would be born with weaker TEOAEs than females in SS twin pairs, as well as show similar TEOAE levels as males in OS t win pairs (masculinized TEOAEs). That expectation was not confirmed here. Contrary, female twins in OS-pairs showed 2.1 dB larger mean TEOAE levels compared to their male co-twins (p = 0.002, n = 76 females), and female twins in OS twin pairs showed numerically larger TEOAEs than females in SS twin pairs, although no statistically significant difference existed (Figure 1). After the publication of the heredity studies (McFadden and Loehlin, 1995; McFadden, et al., 1996), McFadden (2000) has reported that oral contraceptives may produce a weak masculinizing effect on OAEs. Thus, the use of oral contraceptives may have affected the female twins' OAEs differently, as the use cannot be excluded in the group of university students (McFadden and Loehlin, 1995; McFadden, et al., 1996). Moreover, it has been found that TEOAEs can be modulated postnatally by sex hormones, in the form of hormonal treatment, indicating that hormones do influence TEOAEs, although TEOAEs "may not be used as an unequivocal measure of prenatal androgen exposure" (Burke, et al., 2020). Moreover, the neonatal twins only received the prenatal surge of testosterone. The absence of a second surge may have contributed to the lack of masculinization of females in OS twin pairs' TEOAEs, although it is yet unclear why such a twin testosterone transfer effect would be attributed to postnatal hormone exposure.

Cohen's effect sizes for ear and sex differences in TEOAE were similar around 0.2-0.4 for neonatal twins and non-twins, indicating small effects. Similarly, the effect of ear on TEOAEs was 0.24 in young adult twins (McFadden, 2009; McFadden, et al., 1996). In contrast, the effect of sex on TEOAEs in young adult twins was 0.76 (McFadden, 2009; McFadden, et al., 1996), indicating a large effect (0.7 = large). Recordings of auditory evoked potentials in preschoolers, teens and adults support that sex differences in subcortical auditory function emerge throughout development (Krizman, et al., 2019). However, Krizman, et al. (2019) found that some subcortical sex differences emerge well after the cochlea is mature. Here, the TEOAE effect size difference indicates that sex differences may become more pronounced throughout development at the level of the cochlea specifically, perhaps due to hearing maturation and androgens.

It is also noteworthy that females in OS twin pairs demonstrated significantly larger TEOAEs compared to female non-twins (p = .045). We have no explanation for this difference, and it is tempting to discuss androgens in light of the difference, but as the median difference was only 0.9 dB and the female non-twin group was large (n = 10 526 females) there is a high risk for a false positive finding. The same applies to the significant difference observed in TEOAE level between the twins and non-twins (p = .045), where the non-twin group was even larger (n = 21 199), and the median TEOAE difference was only 0.6 dB (Table 1) (or 0.4 dB based on the statistical analysis with the twin pair average).

The heredity estimates are somewhat larger here when using Falconer's formula, compared to the estimates of McFadden, et al. (1996). Heritability estimates are generally not associated with great precision, due to large standard errors. However, the closer the within-pair relationship under comparison (i.e., H2 closer to 1), and the larger the sample size, the more precise the estimate (Falconer and Mackay, 1996). It is with caution we demonstrate a 100% heredity based on the mean TEOAE, due to the uncertainties in the estimates. When primarily studying the estimated MZ correlation for the mean TEOAE of r = 0.77, and the MZ intra-twin pair correlations of r = 0.51-0.68 for various ear combinations one may argue that the heredity may be in the order of 75% as previously suggested (Table 5). However, the bootstrapping analysis revealed that 75% of all estimates of H2 exceeded 69% heritability, which suggests TEOAE heritability may be larger based on neonatal TEOAEs. Furthermore , the Cohen's effect size was notably large at 0.74 for the estimated DZ compared to MZ intra-twin pair correlations, as 0.5 indicates a large effect size.

Table 5. Estimated intra-twin pair correlation coefficients for monozygotic (MZ, estimated n = 75) and dizygotic (DZ, n = 76) twin pairs. The analysis included both left and right ears for the L1-R2 + R1-L2 comparison (MZ, estimated n = 150 ears; DZ, n = 152 ears). The differences between correlation coefficients was calculated using Fisher's r-to-z transform with a two-tailed t-test.

Twin 1 vs. Twin 2 MZ DZ rMZ vs. rDZ
r r p
L-L .62 .30 .01
R-R .51 .24 .06
L1-R2 + R1-L2 .68 .19 .0001
Ear-average TEOAE- Ear-average TEOAE .77 .27 < .0001
L=left, R=right. Twin 1 was born before Twin 2.


Differences in the TEOAE method and criteria used between this study and that of McFadden, et al. (1996) influenced the TEOAE levels in the two studies. In this study, a non-linear mode with 3 ms onset was used, whereas a linear averaging starting at 6 ms was used by McFadden, et al. (1996), i.e., frequency components above approximately 3 kHz will most probably be omitted (Keefe, 2012; Kemp, 1986). Furthermore, we used a non-linear mode with a 20 dB larger stimulus level than that of McFadden, et al. (1996) (82 dB SPL peak vs. about 62 dB SPL peak). The relatively low stimulus level in combination with linear averaging used by McFadden, et al. (1996) might enhance the possibility to detect small TEOAE differences (cf. quinine-induced changes in TEOAE threshold, (Berninger, et al., 1998; Karlsson, et al., 1995)). Consequently, the TEOAE level was about 8 dB larger in the present study, reflecting both the differences in method, and age of the subjects (TEOAEs are up to 10 dB larger i n neonates than in adults (Ferguson, et al., 2000; Kemp, et al., 1990)).

Intra-twin pair correlations (Pearson's r) were used in this study, and by McFadden, et al. (1996) to estimate heredity. McFadden, et al. (1996) used same-ear comparisons to estimate heredity, and not opposite ears. Unexpectedly, our results indicate that the estimated heredity may be larger for opposite ears than for same ears. Interestingly, the largest intra-twin pair correlation displayed by McFadden, et al. (1996) was for opposite ears for MZ male twin pairs (r = 0.83), although the overall difference between same ears and opposite ears were less evident than in the present study. The difference may be explained by mirror-image twinning, i.e., that a right-side feature in one MZ twin appear asymmetrically, on the left side, of the other MZ twin. Mirror-image twinning occurs in about 10-15% of MZ twins that are otherwise healthy, a result of late zygotic splitting (Hall, 2003; McNamara, et al., 2016). Mirror-image features can even exist in as many as 25% of MZ twins, as refl ected by eye and ear defects (McNamara, et al., 2016; Springer and Searleman, 1978). The young adult MZ group also revealed features of mirror-imaging, as four pairs were reported to have opposite handedness (and footedness) (McFadden and Loehlin, 1995; McFadden, et al., 1996). McFadden and Loehlin (1995) also considered the possibility of similar SOAE frequencies being inherited by MZ co-twins, which is another form of mirroring, that may be reflected in our high heritability estimates. The intra-twin pair correlations did not differ between same-ear (L-L and R-R) and opposite-ear (L1-R2 + R1-L2) comparisons for OS and SS twin pairs (Table 2), or for random pairs (Table 3), and not significantly for estimated DZ or estimated MZ correlation coefficients (Table 5). The mirror-image effect, reflected in comparably large opposite-ear correlations for MZ twins, is particularly noticeable in the heredity estimates, when the estimated MZ correlation coefficients are compared to the co rrelation coefficients of DZ twins (Table 5). Consequently, we believe that the intra-twin pair correlations based on the ear-average TEOAE (Table 5) is the most appropriate to interpret in the estimation of heredity, as the risk that the mirror-image effect becoming a confounding variable is removed in the ear-average.

Intra-twin pair correlations for same ears of neonatal and young adult DZ twins used to estimate heritability were similar in size: r = .24, and r = .30 for right and left ears respectively (estimated n = 76 pairs), compared to r = .13, and r = .31 for right and left ears, respectively (n = 28 pairs; Table 10 by McFadden et al. (1996)). It should be noted that we estimated heredity based on OS DZ twin correlations, whereas McFadden et al. (1996) used SS DZ twin correlations, but the OS DZ correlations were also similar: r = .10, and r = .47 for right and left ears respectively (n = 17 pairs; Table 4 by McFadden, et al. (1996)). Likewise, the estimated within-pair MZ correlations were also similar to that of MZ young adult twin-pairs: r = .62, and r = .51 for right and left ears respectively (estimated n = 75 pairs), compared to r = .76, and r = .69 for right and left ears respectively (n = 38 pairs; Table 10 by McFadden, et al. (1996)). The MZ young adult twin pairs displayed numerically larger correlation coefficients for left and right ears as compared to the estimates for neonates, but no statistical difference existed (ps > .17, Fisher's r to z transform). Unfortunately the ear-average TEOAE intra-twin pair correlations, used primarily in the heritability estimation due to potential mirroring, could not be compared to previous studies as these type of correlations were not presented (McFadden, et al., 1996), nor the variability in the heritability estimate. Furthermore, TEOAE intra-twin pair comparisons by ear and sex for MZ twins could not be compared, nor data for a SS DZ group, as the estimated MZ correlations were based on the data from the SS and OS twin sets in the present study.

An advantage of studying TEOAEs and heredity in newborn twins is minimal environmental influence, e.g. differences in noise exposure within a twin pair, which is a difference to previous heredity and OAE studies, wherein students were recruited at a university (McFadden and Loehlin, 1995; McFadden, et al., 1996). Another factor that may have an effect later in development is sex hormones (e.g., (Al-Mana, et al., 2010; Burke, et al., 2020)), as described previously. Another factor that may have modulated adult twin's TEOAEs is cortical activity and selective attention. In animal models it has been found that cortical activity can influence olivocochlear efferents (Dragicevic, et al., 2015), and DPOAEs (Jäger and Kössl, 2016). Specifically, in humans selective auditory and visual attention has been demonstrated to affect DPOAEs (Srinivasan, et al., 2014; Wittekindt, et al., 2014), and may affect a nonlinear version of SFOAEs in humans (Walsh, et al., 2015), but has not been reproduce d for SFOAEs (Beim, et al., 2018; Beim, et al., 2019). Specific studies of TEOAEs and selective attention is lacking, but the results represent another factor that may influence adults, whereas these higher functions have not yet developed in neonates.

Race/ethnicity may also influence TEOAE levels, and specifically sex differences (McFadden, et al., 2018), possibly due to melanocytes in the cochlea (Lin, et al., 2012). Thus, the pigmentation effect on TEOAEs may have differed between this study compared to previous studies, although comparisons are not possible as our ethical consent did not include the collection of this information.

Zygosity estimation represent another difference between this study and that of McFadden, et al. (1996). The methods used are not fully comparable, as we used mathematical estimations of heredity based on the TEOAEs of the OS and SS group, whereas McFadden, et al. (1996) used a questionnaire procedure (Nichols and Bilbro, 1966), with approximately 90% accuracy in identifying zygosity (McFadden and Loehlin, 1995). For future research of OAE heredity, it would be of value to use a different method for determining zygosity in neonatal twins to confirm a large heredity contribution, preferably by analyzing DNA samples. Another difference between the studies was the DZ group under comparison. Estimated MZ intra-twin pair correlations were compared to DZ twins of OS here, whereas McFadden, et al. (1996) used exclusively DZ twins of SS. The DZ twins of OS were excluded by McFadden, et al. (1996) due to the previous research showing weaker OAEs for the DZ females in OS-pairs (McFadden, 1993a ), which we did not observe in our data (Figure 1). A factor that could potentially influence our comparison of SS and OS twins, and perhaps that we use a DZ group consisting of solely OS-pairs, were the sex differences in TEOAE. Thus, we analyzed randomly paired twins under the condition that their randomly assigned co-twin was of the same sex and from the same group (SS or OS) as their biological co-twin. Although a trend can be perceived of a larger variability for the OS-group (than for the SS-group) when comparing different ear conditions, no significant difference was found when comparing the SS- and OS-group of randomly paired twins' TEOAE correlations (Table 3). Moreover, the correlation coefficients could not be distinguished from zero for any group or any ear comparison (Table 3), indicating that heredity and not sex differences are the cause for the larger correlation coefficients for SS twin pairs, as compared to OS twin pairs (Table 2), which is also reflected in the estimated MZ and DZ correlations (Table 5).

5. Conclusion
In a large consecutive sample of neonatal twins, born over a 6-year period, we demonstrate that individual differences in TEOAE levels are largely inherited. Furthermore, neonatal twins show similar TEOAE lateral asymmetry (right > left) and TEOAE sex differences (female > male) as neonatal non-twins and young adult twins.

The heritability estimates based on ear-average TEOAE levels indicate that up to 100% of the TEOAE level may be inherited, and 75% of our heritability estimates exceeded 69%. Thus, the heritability of the TEOAEs in neonates may be larger than the previous estimate of about 75% percent based on young adult twin's TEOAEs. Further study in neonates with DNA sampling for zygosity would be of value, to confirm whether TEOAEs are partially or fully inherited.

Author statement
All authors contributed to the study design, analysis, and interpretation of data, as well as drafting the article, and the approval of the final manuscript. Erik Berninger and Åke Olofsson also contributed in the data collection. All data are sharable upon request.

Data statement
Data are sharable upon request.

Declaration of Competing Interest
None.

Acknowledgements
We would like to acknowledge all that have contributed to a successful hearing screening program, and the continued research at the Scientific Center for Advanced Pediatric Audiology (SCAPA), Karolinska Institutet, Karolinska University Hospital. Arne Leijon is greatly acknowledged for fruitful discussions on variance estimates for heritability. This study was supported by Oticon Foundation, Tysta Skolan Foundation, the regional agreement on medical training and clinical research (ALF) between Region Stockholm and Karolinska Institutet, Karolinska University Hospital, the Jerring foundation and the Eskilstuna-Huddinge Research Fund. We also thank Swedish Society of Medicine for a travel grant to 31st Midwinter Research Meeting, Association for Research in Otolaryngology, Phoenix, AZ, USA, 2008 where part of this study was presented.

References
Aidan et al., 1997
D. Aidan, P. Lestang, P. Avan, P. Bonfils
Characteristics of transient-evoked otoacoustic emissions (TEOES) in neonates
Acta Otolaryngol, 117 (1997), pp. 25-30
CrossRefView Record in ScopusGoogle Scholar
Al-Mana et al., 2010
D. Al-Mana, B. Ceranic, O. Djahanbakhch, L.M. Luxon
Alteration in auditory function during the ovarian cycle
Hear Res, 268 (2010), pp. 114-122
ArticleDownload PDFView Record in ScopusGoogle Scholar
Alvan et al., 2017
G. Alvan, E. Berninger, L.L. Gustafsson, K.K. Karlsson, G. Paintaud, M. Wakelkamp
Concentration-Response Relationship of Hearing Impairment Caused by Quinine and Salicylate: Pharmacological Similarities but Different Molecular Mechanisms
Basic Clin Pharmacol Toxicol, 120 (2017), pp. 5-13
CrossRefView Record in ScopusGoogle Scholar
Beim et al., 2018
J.A. Beim, A.J. Oxenham, M. Wojtczak
Examining replicability of an otoacoustic measure of cochlear function during selective attention
J Acoust Soc Am, 144 (2018), pp. 2882-2895
CrossRefView Record in ScopusGoogle Scholar
Beim et al., 2019
J.A. Beim, A.J. Oxenham, M. Wojtczak
No effects of attention or visual perceptual load on cochlear function, as measured with stimulus-frequency otoacoustic emissions
J Acoust Soc Am, 146 (2019), p. 1475
CrossRefView Record in ScopusGoogle Scholar
Berninger, 2007
E. Berninger
Characteristics of normal newborn transient-evoked otoacoustic emissions: ear asymmetries and sex effects
Int J Audiol, 46 (2007), pp. 661-669
CrossRefView Record in ScopusGoogle Scholar
Berninger, 2014
E. Berninger
Letter to the Editor regarding "Otoacoustic emissions in newborn hearing screening: a systematic review of the effects of different protocols on test outcomes"
Int J Pediatr Otorhinolaryngol, 78 (2014), p. 2022
ArticleDownload PDFView Record in ScopusGoogle Scholar
Berninger and Westling, 2011
E. Berninger, B. Westling
Outcome of a universal newborn hearing-screening programme based on multiple transient-evoked otoacoustic emissions and clinical brainstem response audiometry
Acta Otolaryngol, 131 (2011), pp. 728-739
CrossRefView Record in ScopusGoogle Scholar
Berninger et al., 1998
E. Berninger, K.K. Karlsson, G. Alvan
Quinine reduces the dynamic range of the human auditory system
Acta Otolaryngol, 118 (1998), pp. 46-51
View Record in ScopusGoogle Scholar
Berninger et al., 1995
E. Berninger, K.K. Karlsson, U. Hellgren, G. Eskilsson
Magnitude changes in transient evoked otoacoustic emissions and high-level 2f1-f2 distortion products in man during quinine administration
Scand Audiol, 24 (1995), pp. 27-32
View Record in ScopusGoogle Scholar
Bray and Kemp, 1987
P. Bray, D. Kemp
An advanced cochlear echo technique suitable for infant screening
Br J Audiol, 21 (1987), pp. 191-204
CrossRefView Record in ScopusGoogle Scholar
Burke et al., 2020
S.M. Burke, J.O. van Heesewijk, W.M. Menks, D.T. Klink, B.P.C. Kreukels, P.T. Cohen-Kettenis, J. Bakker
Postnatal Effects of Sex Hormones on Click-Evoked Otoacoustic Emissions: A Study of Adolescents with Gender Dysphoria
Arch Sex Behav, 49 (2020), pp. 455-465
CrossRefView Record in ScopusGoogle Scholar
Cassidy and Ditty, 2001
J.W. Cassidy, K.M. Ditty
Gender differences among newborns on a transient otoacoustic emissions test for hearing
J Music Ther, 38 (2001), pp. 28-35
CrossRefView Record in ScopusGoogle Scholar
Cheatham et al., 2014
M.A. Cheatham, R.J. Goodyear, K. Homma, P.K. Legan, J. Korchagina, S. Naskar, J.H. Siegel, P. Dallos, J. Zheng, G.P. Richardson
Loss of the tectorial membrane protein CEACAM16 enhances spontaneous, stimulus-frequency, and transiently evoked otoacoustic emissions
J Neurosci, 34 (2014), pp. 10325-10338
CrossRefView Record in ScopusGoogle Scholar
Clemens et al., 1978
L.G. Clemens, B.A. Gladue, L.P. Coniglio
Prenatal endogenous androgenic influences on masculine sexual behavior and genital morphology in male and female rats
Horm Behav, 10 (1978), pp. 40-53
ArticleDownload PDFView Record in ScopusGoogle Scholar
Cohen, 1992
J. Cohen
A power primer
Psychological bulletin, 112 (1992), pp. 155-159
CrossRefView Record in ScopusGoogle Scholar
Dragicevic et al., 2015
C.D. Dragicevic, C. Aedo, A. Leon, M. Bowen, N. Jara, G. Terreros, L. Robles, P.H. Delano
The Olivocochlear Reflex Strength and Cochlear Sensitivity are Independently Modulated by Auditory Cortex Microstimulation
J Assoc Res Otolaryngol, 16 (2015), pp. 223-240
CrossRefView Record in ScopusGoogle Scholar
Falconer and Mackay, 1996
D.S. Falconer, T.F.C. Mackay
Introduction to quantitative genetics Longman
Harlow (1996)
Google Scholar
Fellman and Eriksson, 2006
J. Fellman, A.W. Eriksson
Weinberg's differential rule reconsidered
Hum Biol, 78 (2006), pp. 253-275
CrossRefView Record in ScopusGoogle Scholar
Ferguson et al., 2000
M.A. Ferguson, P.A. Smith, A.C. Davis, M.E. Lutman
Transient-evoked otoacoustic emissions in a representative population sample aged 18 to 25 years
Audiology, 39 (2000), pp. 125-134
CrossRefView Record in ScopusGoogle Scholar
Franklin et al., 1992
D.J. Franklin, M.J. McCoy, G.K. Martin, B.L. Lonsbury-Martin
Test/retest reliability of distortion-product and transiently evoked otoacoustic emissions
Ear Hear, 13 (1992), pp. 417-429
CrossRefView Record in ScopusGoogle Scholar
Guinan, 2006
J.J. Guinan Jr.
Olivocochlear efferents: anatomy, physiology, function, and the measurement of efferent effects in humans
Ear Hear, 27 (2006), pp. 589-607
CrossRefView Record in ScopusGoogle Scholar
Guinan, 2018
J.J. Guinan Jr.
Olivocochlear efferents: Their action, effects, measurement and uses, and the impact of the new conception of cochlear mechanical responses
Hear Res, 362 (2018), pp. 38-47
ArticleDownload PDFView Record in ScopusGoogle Scholar
Hall, 2003
J.G. Hall
Twinning
Lancet, 362 (2003), pp. 735-743
ArticleDownload PDFView Record in ScopusGoogle Scholar
Harris et al., 1991
F.P. Harris, R. Probst, R. Wenger
Repeatability of transiently evoked otoacoustic emissions in normally hearing humans
Audiology, 30 (1991), pp. 135-141
CrossRefView Record in ScopusGoogle Scholar
Hines et al., 2015
M. Hines, M. Constantinescu, D. Spencer
Early androgen exposure and human gender development
Biology of sex differences, 6 (2015), p. 3
Google Scholar
Johnsen and Elberling, 1982a
N.J. Johnsen, C. Elberling
Evoked acoustic emissions from the human ear
II. Normative data in young adults and influence of posture. Scand Audiol, 11 (1982), pp. 69-77
CrossRefView Record in ScopusGoogle Scholar
Johnsen and Elberling, 1982b
N.J. Johnsen, C. Elberling
Evoked acoustic emissions from the human ear
I. Equipment and response parameters. Scand Audiol, 11 (1982), pp. 3-12
CrossRefView Record in ScopusGoogle Scholar
Johnsen et al., 1988
N.J. Johnsen, P. Bagi, J. Parbo, C. Elberling
Evoked acoustic emissions from the human ear. IV. Final results in 100 neonates
Scand Audiol, 17 (1988), pp. 27-34
View Record in ScopusGoogle Scholar
Jäger and Kössl, 2016
K. Jäger, M. Kössl
Corticofugal Modulation of DPOAEs in Gerbils
Hear Res, 332 (2016), pp. 61-72
ArticleDownload PDFView Record in ScopusGoogle Scholar
Karlsson et al., 1991
K.K. Karlsson, E. Berninger, G. Alvan
The effect of quinine on psychoacoustic tuning curves, stapedius reflexes and evoked otoacoustic emissions in healthy volunteers
Scand Audiol, 20 (1991), pp. 83-90
CrossRefView Record in ScopusGoogle Scholar
Karlsson et al., 1995
K.K. Karlsson, E. Berninger, L.L. Gustafsson, G. Alvan
Pronounced quinine-induced cochlear hearing loss. A mechanistic study in one volunteer at multiple stable plasma concentrations
Journal of Audiological Medicine, 4 (1995), pp. 12-24
View Record in ScopusGoogle Scholar
Keefe, 2012
D.H. Keefe
Moments of click-evoked otoacoustic emissions in human ears: group delay and spread, instantaneous frequency and bandwidth
J Acoust Soc Am, 132 (2012), pp. 3319-3350
CrossRefView Record in ScopusGoogle Scholar
Kei et al., 1997
J. Kei, B. McPherson, V. Smyth, S. Latham, J. Loscher
Transient evoked otoacoustic emissions in infants: effects of gender, ear asymmetry and activity status
Audiology, 36 (1997), pp. 61-71
CrossRefView Record in ScopusGoogle Scholar
Kemp, 1978
D.T. Kemp
Stimulated acoustic emissions from within the human auditory system
J Acoust Soc Am, 64 (1978), pp. 1386-1391
CrossRefView Record in ScopusGoogle Scholar
Kemp, 1986
D.T. Kemp
Otoacoustic emissions, travelling waves and cochlear mechanisms
Hear Res, 22 (1986), pp. 95-104
ArticleDownload PDFView Record in ScopusGoogle Scholar
Kemp et al., 1990
D.T. Kemp, S. Ryan, P. Bray
A guide to the effective use of otoacoustic emissions
Ear Hear, 11 (1990), pp. 93-105
CrossRefView Record in ScopusGoogle Scholar
Khalfa et al., 1997
S. Khalfa, T. Morlet, C. Micheyl, A. Morgon, L. Collet
Evidence of peripheral hearing asymmetry in humans: clinical implications
Acta Otolaryngol, 117 (1997), pp. 192-196
CrossRefView Record in ScopusGoogle Scholar
Kinsley et al., 1986
C. Kinsley, J. Miele, C. Konen, L. Ghiraldi, B. Svare
Intrauterine contiguity influences regulatory activity in adult female and male mice
Horm Behav, 20 (1986), pp. 7-12
ArticleDownload PDFView Record in ScopusGoogle Scholar
Krizman et al., 2019
J. Krizman, S. Bonacina, N. Kraus
Sex differences in subcortical auditory processing emerge across development
Hear Res, 380 (2019), pp. 166-174
ArticleDownload PDFView Record in ScopusGoogle Scholar
Lamminmäki et al., 2012
A. Lamminmäki, M. Hines, T. Kuiri-Hänninen, L. Kilpeläinen, L. Dunkel, U. Sankilampi
Testosterone measured in infancy predicts subsequent sex-typed behavior in boys and in girls
Hormones and Behavior, 61 (2012), pp. 611-616
ArticleDownload PDFView Record in ScopusGoogle Scholar
Lin et al., 2012
F.R. Lin, P. Maas, W. Chien, J.P. Carey, L. Ferrucci, R. Thorpe
Association of skin color, race/ethnicity, and hearing loss among adults in the USA
J Assoc Res Otolaryngol, 13 (2012), pp. 109-117
CrossRefView Record in ScopusGoogle Scholar
Marshall and Heller, 1996
L. Marshall, L.M. Heller
Reliability of transient-evoked otoacoustic emissions
Ear Hear, 17 (1996), pp. 237-254
View Record in ScopusGoogle Scholar
McFadden, 1993a
D. McFadden
A masculinizing effect on the auditory systems of human females having male co-twins
Proc Natl Acad Sci U S A, 90 (1993), pp. 11900-11904
CrossRefView Record in ScopusGoogle Scholar
McFadden, 1993b
D. McFadden
A speculation about the parallel ear asymmetries and sex differences in hearing sensitivity and otoacoustic emissions
Hear Res, 68 (1993), pp. 143-151
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden, 2000
D. McFadden
Masculinizing effects on otoacoustic emissions and auditory evoked potentials in women using oral contraceptives
Hear Res, 142 (2000), pp. 23-33
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden, 2008
D. McFadden
What Do Sex, Twins, Spotted Hyenas, ADHD, and Sexual Orientation Have in Common?
Perspectives on psychological science: a journal of the Association for Psychological Science, 3 (2008), pp. 309-323
CrossRefView Record in ScopusGoogle Scholar
McFadden, 2009
D. McFadden
Masculinization of the mammalian cochlea
Hear Res, 252 (2009), pp. 37-48
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden and Loehlin, 1995
D. McFadden, J.C. Loehlin
On the heritability of spontaneous otoacoustic emissions: A twins study
Hear Res, 85 (1995), pp. 181-198
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden et al., 1996
D. McFadden, J.C. Loehlin, E.G. Pasanen
Additional findings on heritability and prenatal masculinization of cochlear mechanisms: click-evoked otoacoustic emissions
Hear Res, 97 (1996), pp. 102-119
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden et al., 2006a
D. McFadden, E.G. Pasanen, J. Raper, H.S. Lange, K. Wallen
Sex differences in otoacoustic emissions measured in rhesus monkeys (Macaca mulatta)
Horm Behav, 50 (2006), pp. 274-284
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden et al., 2006b
D. McFadden, E.G. Pasanen, M.L. Weldele, S.E. Glickman, N.J. Place
Masculinized otoacoustic emissions in female spotted hyenas (Crocuta crocuta)
Horm Behav, 50 (2006), pp. 285-292
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden et al., 2009
D. McFadden, E.G. Pasanen, M.D. Valero, E.K. Roberts, T.M. Lee
Effect of prenatal androgens on click-evoked otoacoustic emissions in male and female sheep (Ovis aries)
Horm Behav, 55 (2009), pp. 98-105
ArticleDownload PDFView Record in ScopusGoogle Scholar
McFadden et al., 2018
D. McFadden, E.G. Pasanen, M.M. Maloney, E.M. Leshikar, M.H. Pho
Correlations between otoacoustic emissions and performance in common psychoacoustical tasks
J Acoust Soc Am, 143 (2018), p. 2355
CrossRefView Record in ScopusGoogle Scholar
McNamara et al., 2016
H.C. McNamara, S.C. Kane, J.M. Craig, R.V. Short, M.P. Umstad
A review of the mechanisms and evidence for typical and atypical twinning
Am J Obstet Gynecol, 214 (2016), pp. 172-191
ArticleDownload PDFView Record in ScopusGoogle Scholar
Nichols and Bilbro, 1966
R.C. Nichols, W.C. Bilbro Jr.
The Diagnosis of Twin Zygosity
Hum Hered, 16 (1966), pp. 265-275
CrossRefView Record in ScopusGoogle Scholar
Ren and He, 2020
T. Ren, W. He
Two-tone distortion in reticular lamina vibration of the living cochlea
Commun Biol, 3 (2020), p. 35
CrossRefView Record in ScopusGoogle Scholar
Shera and Guinan, 1999
C.A. Shera, J.J. Guinan Jr.
Evoked otoacoustic emissions arise by two fundamentally different mechanisms: a taxonomy for mammalian OAEs
J Acoust Soc Am, 105 (1999), pp. 782-798
View Record in ScopusGoogle Scholar
Springer and Searleman, 1978
S. Springer, A. Searleman
Laterality in twins: The relationship between handedness and hemispheric asymmetry for speech
Behav Genet, 8 (1978), pp. 349-357
View Record in ScopusGoogle Scholar
Srinivasan et al., 2014
S. Srinivasan, A. Keil, K. Stratis, A.F. Osborne, C. Cerwonka, J. Wong, B.L. Rieger, V. Polcz, D.W. Smith
Interaural attention modulates outer hair cell function
Eur J Neurosci, 40 (2014), pp. 3785-3792
CrossRefView Record in ScopusGoogle Scholar
Thornton et al., 2003
A.R. Thornton, N. Marotta, C.R. Kennedy
The order of testing effect in otoacoustic emissions and its consequences for sex and ear differences in neonates
Hear Res, 184 (2003), pp. 123-130
ArticleDownload PDFView Record in ScopusGoogle Scholar
Walsh et al., 2015
K.P. Walsh, E.G. Pasanen, D. McFadden
Changes in otoacoustic emissions during selective auditory and visual attention
J Acoust Soc Am, 137 (2015), pp. 2737-2757
CrossRefView Record in ScopusGoogle Scholar
Veuillet et al., 1991
E. Veuillet, L. Collet, R. Duclaux
Effect of contralateral acoustic stimulation on active cochlear micromechanical properties in human subjects: dependence on stimulus variables
J Neurophysiol, 65 (1991), pp. 724-735
CrossRefView Record in ScopusGoogle Scholar
Wittekindt et al., 2014
A. Wittekindt, J. Kaiser, C. Abel
Attentional modulation of the inner ear: a combined otoacoustic emission and EEG study
J Neurosci, 34 (2014), pp. 9995-10002
CrossRefView Record in ScopusGoogle Scholar
vom Saal, 1989
F.S. vom Saal
Sexual differentiation in litter-bearing mammals: influence of sex of adjacent fetuses in utero
Journal of animal science, 67 (1989), pp. 1824-1840
View Record in ScopusGoogle Scholar
vom Saal and Bronson, 1980
F.S. vom Saal, F.H. Bronson
Sexual characteristics of adult female mice are correlated with their blood testosterone levels during prenatal development
Science, 208 (1980), pp. 597-599
View Record in ScopusGoogle Scholar
View Abstract
© 2020 The Author(s). Published by Elsevier B.V.

1-s2.0-S0378595520X00104-cov150h.gif

Publication date: Available online 28 October 2020

Source: Hearing Research

Author(s): Marlin Johansson, Åke Olofsson, Erik Berninger

View on the web

Αναζήτηση αυτού του ιστολογίου

! # Ola via Alexandros G.Sfakianakis on Inoreader